1
|
Teng Y, Xue H, Deng X, Luo Y, Wu T. The role of phosphatidylethanolamine-binding protein (PEBP) family in various diseases: Mechanisms and therapeutic potential. PROGRESS IN BIOPHYSICS AND MOLECULAR BIOLOGY 2025; 196:102-113. [PMID: 40220872 DOI: 10.1016/j.pbiomolbio.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/27/2025] [Accepted: 04/09/2025] [Indexed: 04/14/2025]
Abstract
This article focuses on the phosphatidylethanolamine-binding protein (PEBP) family proteins, detailing PEBP1 and PEBP4 due to limited information on PEBP2 and PEBP3, in cellular signaling pathways and research in a spectrum of pathologies, including diverse cancers, metabolic disorders, immunological diseases and a subset of organ-specific diseases. It outlines the mechanisms through which PEBP1 and PEBP4 regulate essential signaling pathways that are critical for cellular processes such as proliferation, apoptosis, and metastasis. Recent advancements have shown further understanding of these proteins' roles in pathophysiology and their potential as future therapeutic targets. The findings suggest that the impact of PEBP1 and PEBP4 on the course of different diseases has underscored their potential for more in-depth medical research and novel clinically targeted therapies.
Collapse
Affiliation(s)
- Yeying Teng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Haiping Xue
- Industrial Development Center, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Xiaoliang Deng
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yanqun Luo
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tao Wu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China.
| |
Collapse
|
2
|
Yu W, Wang Y, Han J, Hu D, Zhao C, Liu Z, Wu Z. Capsular Polysaccharides Are Crucial for the Biosorption of Aqueous Pb(II) Ions by Limosilactobacillus fermentum B44 In Vitro. Biol Trace Elem Res 2025:10.1007/s12011-025-04608-y. [PMID: 40208435 DOI: 10.1007/s12011-025-04608-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/31/2025] [Indexed: 04/11/2025]
Abstract
Lead (Pb) is a widespread heavy metal pollutant that presents substantial risks to ecological systems and human health. The utilization of probiotic lactic acid bacteria (LAB) for the detoxification of Pb(II) in the host has aroused great interest. However, few studies had focused on the roles of capsular polysaccharide (CPS) in the bacteria adsorption of Pb(II). In the present study, L. fermentum B44 was selected out of nine lactobacilli for its superior absorption capacity of Pb(II) in vitro. The adsorbed Pb(II) ions by B44 cells were predominantly enriched on the capsule. Removal of the CPS from cells of strain B44 by 1 mol/L guanidine hydrochloride (GuHCl) would decrease the adsorption capacity of the host cells. The CPS showed a high capacity for Pb(II) adsorption, and its adsorption behaviour was more accurately described by the Langmuir isotherm model (R2 = 0.97) than by the Freundlich isotherm model (R2 = 0.907). Adsorption of Pb(II) ions would lead the CPS transforming from stacked, porous flakes to rough, condensed ones, with Pb(II) sediment on the CPS surface. Amide groups in B44 CPS were crucial in the adsorption of Pb(II). Our results provided intuitive evidence of the Pb(II) adsorption site on B44 cells and the protection of CPS to the bacterial cells.
Collapse
Affiliation(s)
- Wenhui Yu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
- College of Food Sciences & Technology, Shanghai Ocean University, Shanghai, 201306, China
| | - Yitian Wang
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
- State Key Laboratory of Microbial Metabolism, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Shanghai, 200240, China
| | - Jin Han
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
| | - Dan Hu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
| | - Chenbing Zhao
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China
| | - Zhengjun Wu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Dairy Research Institute, Bright Dairy & Food Co., Ltd, Shanghai, 201103, China.
| |
Collapse
|
3
|
Fan X, Song Y, Liu Y, Song J, Zeng J, Li Z, Xu J, Xue C. Effects of mitochondrial lipidome alterations on quality deterioration of Larimichthys crocea postmortem storage: New insight from the perspective of mediating mitochondria-dependent apoptosis. Food Chem 2025; 468:142461. [PMID: 39693887 DOI: 10.1016/j.foodchem.2024.142461] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 11/27/2024] [Accepted: 12/10/2024] [Indexed: 12/20/2024]
Abstract
Apoptosis occurs in the myocyte of fish postmortem storage. Based on the important role of mitochondrial lipid molecules in regulating apoptosis, the study aims to investigate the potential impact of mitochondrial lipids on apoptosis and quality deterioration of large yellow croaker. A total of 1079 lipid molecule species in 13 classes were identified in mitochondria. PC and PE decreased by 17.40 % and 28.31 % at 24 h, which induces mitochondrial damage and induces oxidative stress. Cytochrome c induced CL oxidation mediated by ROS (Oxidized CL increased by 30.65 %), resulting in cytochrome c release and activates caspase-3. The cytochrome c of cytoplasm and caspase-3 activity increased by 79.32 % and 82.72 % from 0 to 24 h, which led to significant apoptosis. Accumulation of ROS and activated caspase-3 during apoptosis induced muscle oxidation and softening. These findings provide new insights into the relationship between mitochondrial lipid changes and apoptosis and quality deterioration in fish postmortem storage.
Collapse
Affiliation(s)
- Xiaowei Fan
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China
| | - Yu Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China.
| | - Yanjun Liu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China.
| | - Junyi Song
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China
| | - Junpeng Zeng
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China
| | - Zhaojie Li
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China.
| | - Jie Xu
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China.
| | - Changhu Xue
- State Key Laboratory of Marine Food Processing & Safety Control, College of Food Science and Engineering, Ocean University of China, No. 1299, Sansha Road, Qingdao, Shandong Province 266404, China; Qingdao Marine Science and Technology Center, Qingdao, Shandong Province 266235, China.
| |
Collapse
|
4
|
Sun JY, Qi SJ, Chen Q, Liu KX, Liu HY, Zheng HB, Sun B, Lou HX. Design, Synthesis, and Biological Evaluation of Marchantin C-NO Donor Hybrids for Overcoming Pgp-Mediated Drug Resistance by Targeting Lysosome. J Med Chem 2025; 68:5503-5528. [PMID: 40014032 DOI: 10.1021/acs.jmedchem.4c02733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2025]
Abstract
A series of marchantin C-NO donor hybrids were designed, synthesized, and evaluated for their antitumor activity in vitro and in vivo. Notably, MC-furoxan hybrid 14 exhibited the best selective inhibitory activity against MCF-7/ADR (IC50 = 0.024 μM) with 883 times potency compared with MCF-7 cells (IC50 = 21.20 μM), and the cytotoxicity toward A549/Taxol (IC50 = 1.43 μM) increased 17-fold compared with that in A549 cells (IC50 = 23.75 μM). Preliminary pharmacological studies revealed that 14 could "hijack" the lysosomal Pgp and release NO to produce reactive oxygen species (ROS) in lysosomes, resulting in lysosomal membrane permeabilization (LMP) and potentiated cytotoxicity. Additionally, compound 14 achieved stronger antitumor activity and superior biosafety at relatively low doses than paclitaxel in the A549/Taxol xenograft model. In summary, this study provides a promising strategy for the design of such MC-furoxan hybrids like 14 to overcome MDR via the utilization of lysosomal Pgp transport activity.
Collapse
Affiliation(s)
- Jia-Yu Sun
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Si-Jie Qi
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Qian Chen
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Ke-Xin Liu
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Hao-Yu Liu
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Hong-Bo Zheng
- Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, P. R. China
| | - Bin Sun
- National Glycoengineering Research Center, Shandong University, Qingdao 266237, P. R. China
| | - Hong-Xiang Lou
- Key Laboratory of Natural Products & Chemical Biology, Ministry of Education, Shandong University, Jinan 250012, P. R. China
| |
Collapse
|
5
|
Zhao Y, Lin S, Zeng W, Lin X, Qin X, Miu B, Gao S, Wu H, Liu J, Chen X. JS-K activates G2/M checkpoints through the DNA damage response and induces autophagy via CAMKKβ/AMPKα/mTOR pathway in bladder cancer cells. J Cancer 2024; 15:343-355. [PMID: 38169515 PMCID: PMC10758033 DOI: 10.7150/jca.86393] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Accepted: 08/21/2023] [Indexed: 01/05/2024] Open
Abstract
The aim of this study was to investigate the effects of JS-K, a nitric oxide donor prodrug, on DNA damage and autophagy in bladder cancer (BCa) cells and to explore the potential related mechanisms. Through detecting proliferation viability, cell morphology observation and colony formation assay low concentrations of JS-K significantly inhibited BCa growth while having no effect on normal cells. JS-K induced an increase in the level of DNA damage protein γH2AX and a decrease in the level of DNA damage repair-related proteins PCNA and RAD51 in BCa cells, indicating that JS-K can induce DNA damage in BCa cells and inhibit DNA damage repair. JS-K induced G2/M phase block and calcium overload using flow cytometry analysis. Moreover, we also investigated the levels of cell G2/M cycle checkpoint-related protein and autophagy-associated protein by western blot. The results of our study demonstrated that JS-K induced BCa cells G2/M phase arrest due to upregulating proteins related to DNA damage-related G2/M checkpoint activation (p-ATM, p-ATR, p-Chk1, p-Chk2, and p-Cdc2) and down-regulation of Cyclin B1 protein. In addition, our study demonstrated that JS-K-induced autophagy in BCa cells was related to the CAMKKβ/AMPKα/mTOR pathway.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Xiaojun Chen
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
6
|
Yang H, Mo M, Yang L, Yu J, Li J, Cheng S, Sun B, Xu B, Zhang A, Luo H. A Novel Quinazoline Derivative Prevents and Treats Arsenic-Induced Liver Injury by Regulating the Expression of RecQ Family Helicase. Int J Mol Sci 2023; 24:15521. [PMID: 37958505 PMCID: PMC10647758 DOI: 10.3390/ijms242115521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 10/19/2023] [Accepted: 10/21/2023] [Indexed: 11/15/2023] Open
Abstract
Arsenic is a carcinogenic metalloid toxicant widely found in the natural environment. Acute or prolonged exposure to arsenic causes a series of damages to the organs, mainly the liver, such as hepatomegaly, liver fibrosis, cirrhosis, and even hepatocellular carcinoma. Therefore, it is imperative to seek drugs to prevent arsenic-induced liver injury. Quinazolines are a class of nitrogen heterocyclic compounds with biological and pharmacological effects in vivo and in vitro. This study was designed to investigate the ameliorating effects of quinazoline derivatives on arsenic-induced liver injury and its molecular mechanism. We investigated the mechanism of the quinazoline derivative KZL-047 in preventing and ameliorating arsenic-induced liver injury in vitro by cell cycle and apoptosis. We performed real-time fluorescence quantitative polymerase chain reaction (qPCR) and Western blotting combined with molecular docking. In vivo, the experiments were performed to investigate the mechanism of KZL-047 in preventing and ameliorating arsenic-induced liver injury using arsenic-infected mice. Physiological and biochemical indices of liver function in mouse serum were measured, histopathological changes in liver tissue were observed, and immunohistochemical staining was used to detect changes in the expression of RecQ-family helicases in mouse liver tissue. The results of in vitro experiments showed that sodium arsenite (SA) inhibited the proliferation of L-02 cells, induced apoptosis, blocked the cell cycle at the G1 phase, and decreased the expression of RecQ family helicase; after KZL-047 treatment in arsenic-induced L-02 cells, the expression of RecQ family helicase was upregulated, and the apoptosis rate was slowed, leading to the restoration of the cell viability level. KZL-047 inhibited arsenic-induced oxidative stress, alleviated oxidative damage and lipid peroxidation in vivo, and ameliorated arsenic toxicity-induced liver injury. KZL-047 restored the expression of RecQ family helicase proteins, which is consistent with the results of in vitro studies. In summary, KZL-047 can be considered a potential candidate for the treatment of arsenic-induced liver injury.
Collapse
Affiliation(s)
- Heping Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China (M.M.); (B.S.)
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
| | - Min Mo
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China (M.M.); (B.S.)
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
| | - Langlang Yang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China (M.M.); (B.S.)
| | - Jia Yu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang 550014, China
| | - Jiao Li
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang 550014, China
| | - Sha Cheng
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang 550014, China
| | - Baofei Sun
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China (M.M.); (B.S.)
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
| | - Bixue Xu
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang 550014, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, Guizhou Medical University, Guiyang 550025, China (M.M.); (B.S.)
| | - Heng Luo
- State Key Laboratory of Functions and Applications of Medicinal Plants, Guizhou Medical University, Guiyang 550014, China; (J.Y.); (S.C.); (B.X.)
- Natural Products Research Center of Guizhou Province, Guizhou Medical University, Guiyang 550014, China
| |
Collapse
|
7
|
Kim TW, Lee HG. 6-Shogaol Overcomes Gefitinib Resistance via ER Stress in Ovarian Cancer Cells. Int J Mol Sci 2023; 24:ijms24032639. [PMID: 36768961 PMCID: PMC9916959 DOI: 10.3390/ijms24032639] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Revised: 01/18/2023] [Accepted: 01/20/2023] [Indexed: 02/03/2023] Open
Abstract
In women, ovary cancer is already the fifth leading cause of mortality worldwide. The use of cancer therapies, such as surgery, radiotherapy, and chemotherapy, may be a powerful anti-cancer therapeutic strategy; however, these therapies still have many problems, including resistance, toxicity, and side effects. Therefore, natural herbal medicine has the potential to be used for cancer therapy because of its low toxicity, fewer side effects, and high success. This study aimed to investigate the anti-cancer effect of 6-shogaol in ovarian cancer cells. 6-shogaol induces ER stress and cell death via the reduction in cell viability, the increase in LDH cytotoxicity, caspase-3 activity, and Ca2+ release, and the upregulation of GRP78, p-PERK, p-eIF2α, ATF-4, CHOP, and DR5. Moreover, 6-shogaol treatment medicates endoplasmic reticulum (ER) stress and cell death by upregulating Nox4 and releasing ROS. The knockdown of Nox4 in ovarian cancer cells inhibits ER stress and cell death by blocking the reduction in cell viability and the enhancement of LDH cytotoxicity, caspase-3 activity, Ca2+, and ROS release. In gefitinib-resistant ovarian cancer cells, A2780R and OVCAR-3R, 6-shogaol/gefitinib overcomes gefitinib resistance by inhibiting EMT phenomena such as the reduction in E-cadherin, and the increase in N-cadherin, vimentin, Slug, and Snail. Therefore, our results suggest that 6-shogaol exerts a potential anti-cancer effect in ovarian cancer and combination treatment with 6-shogaol and gefitinib may provide a novel anti-tumor therapeutic strategy in gefitinib-resistant ovarian cancer.
Collapse
Affiliation(s)
- Tae Woo Kim
- Department of Biopharmaceutical Engineering, Dongguk University-WISE, Gyeongju 38066, Republic of Korea
- Correspondence:
| | - Hee Gu Lee
- Immunotherapy Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon 34141, Republic of Korea
| |
Collapse
|
8
|
Enhanced IRE1α Phosphorylation/Oligomerization-Triggered XBP1 Splicing Contributes to Parkin-Mediated Prevention of SH-SY5Y Cell Death under Nitrosative Stress. Int J Mol Sci 2023; 24:ijms24032017. [PMID: 36768338 PMCID: PMC9917145 DOI: 10.3390/ijms24032017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2022] [Revised: 01/10/2023] [Accepted: 01/17/2023] [Indexed: 01/21/2023] Open
Abstract
Mutations in parkin, a neuroprotective protein, are the predominant cause of autosomal recessive juvenile Parkinson's disease. Neuroinflammation-derived nitrosative stress has been implicated in the etiology of the chronic neurodegeneration. However, the interactions between genetic predisposition and nitrosative stress contributing to the degeneration of dopaminergic (DA) neurons remain incompletely understood. Here, we used the SH-SY5Y neuroblastoma cells to investigate the function of parkin and its pathogenic mutants in relation to cell survival under nitric oxide (NO) exposure. The results showed that overexpression of wild-type parkin protected SH-SY5Y cells from NO-induced apoptosis in a reactive oxygen species-dependent manner. Under nitrosative stress conditions, parkin selectively upregulated the inositol-requiring enzyme 1α/X-box binding protein 1 (IRE1α/XBP1) signaling axis, an unfolded protein response signal through the sensor IRE1α, which controls the splicing of XBP1 mRNA. Inhibition of XBP1 mRNA splicing either by pharmacologically inhibiting IRE1α endoribonuclease activity or by genetically knocking down XBP1 interfered with the protective activity of parkin. Furthermore, pathogenic parkin mutants with a defective protective capacity showed a lower ability to activate the IRE1α/XBP1 signaling. Finally, we demonstrated that IRE1α activity augmented by parkin was possibly mediated through interacting with IRE1α to regulate its phosphorylation/oligomerization processes, whereas mutant parkin diminished its binding to and activation of IRE1α. Thus, these results support a direct link between the protective activity of parkin and the IRE1α/XBP1 pathway in response to nitrosative stress, and mutant parkin disrupts this function.
Collapse
|
9
|
Xu L, Wu X, Liu H, Dong G, Zhan J, Li G, Wang G, Liu T. Effects of combination docetaxel with NO treatment to enhance the anti-nasopharyngeal carcinoma efficiency in vitro and in vivo. Eur J Pharm Sci 2022; 178:106281. [PMID: 35995348 DOI: 10.1016/j.ejps.2022.106281] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 08/05/2022] [Accepted: 08/18/2022] [Indexed: 02/05/2023]
Abstract
Nasopharyngeal carcinoma (NPC) is one of the major causes of death in Southern China. Due to the insidious location of NPC, the therapeutic effect of locoregionally advanced NPC is still unsatisfactory. In this work, to improve the treatment efficiency, combining DOC and JS-K to inhibit NPC cells (HNE-1) in vitro was investigated, as well as its possible mechanisms. Moreover, the in vivo effects of DOC and JS-K combination treatment were also evaluated in a xenograft model with HNE-1 cells. In vitro experiments including cell proliferation, migration ability, apoptosis, and expression levels of apoptosis-associated proteins revealed that the combination of DOC and JS-K was able to enhance antitumor effects. In vivo results further confirmed a significant treatment effect without obvious toxicity on mice. The present work provides a promising idea for the treatment of locally advanced NPC.
Collapse
Affiliation(s)
- Lingling Xu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Xidong Wu
- Department of Drug Safety Evaluation, Jiangxi Testing Center of Medical Instruments, Nanchang, 330029, China
| | - Huiqin Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Guangyuan Dong
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; Shantou University Medical College, Shantou, 515063, China
| | - Jiandong Zhan
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China
| | - Guanxue Li
- Pediatric Critical Care Medicine, Zhujiang Hospital of Southern Medical University, Guangzhou, 510280, China
| | - Guanhai Wang
- School of Pharmacy, Guangdong Medical University, Dongguan, 523808, China.
| | - Tao Liu
- Department of Otolaryngology-Head and Neck Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, China; The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
10
|
The Nitric Oxide Donor [Zn(PipNONO)Cl] Exhibits Antitumor Activity through Inhibition of Epithelial and Endothelial Mesenchymal Transitions. Cancers (Basel) 2022; 14:cancers14174240. [PMID: 36077778 PMCID: PMC9454450 DOI: 10.3390/cancers14174240] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 08/27/2022] [Accepted: 08/28/2022] [Indexed: 11/16/2022] Open
Abstract
Simple Summary Nitric oxide (NO) plays a critical pathophysiological role in cancer by modulating several processes, such as angiogenesis, tumor growth, and metastatic potential. The aim of this study was to characterize the antitumor effects of a novel NO donor, [Zn(PipNONO)Cl], on the processes of epithelial– and endothelial–mesenchymal transitions (EMT and EndMT), known to actively participate in cancer progression. Two tumor cells lines were used in this study: human lung cancer cells (A549) and melanoma cells (A375), alone and co-cultured with human endothelial cells. Our results demonstrate that both tumor and endothelial cells were targets of NO action, which impaired EMT and EndMT functional and molecular features. Further studies are needed to finalize the therapeutic use of the novel NO donor. Abstract Exogenous nitric oxide appears a promising therapeutic approach to control cancer progression. Previously, a nickel-based nonoate, [Ni(SalPipNONO)], inhibited lung cancer cells, along with impairment of angiogenesis. The Zn(II) containing derivatives [Zn(PipNONO)Cl] exhibited a protective effect on vascular endothelium. Here, we have evaluated the antitumor properties of [Zn(PipNONO)Cl] in human lung cancer (A549) and melanoma (A375) cells. Metastasis initiates with the epithelial–mesenchymal transition (EMT) process, consisting of the acquisition of invasive and migratory properties by tumor cells. At not cytotoxic levels, the nonoate significantly impaired A549 and A375 EMT induced by transforming growth factor-β1 (TGF-β1). Reduction of the mesenchymal marker vimentin, upregulated by TGF-β1, and restoration of the epithelial marker E-cadherin, reduced by TGF-β1, were detected in both tumor cell lines in the presence of Zn-nonoate. Further, the endothelial–mesenchymal transition achieved in a tumor-endothelial cell co-culture was assessed. Endothelial cells co-cultured with A549 or A375 acquired a mesenchymal phenotype with increased vimentin, alpha smooth muscle actin and Smad2/3, and reduced VE-cadherin. The presence of [Zn(PipNONO)Cl] maintained a typical endothelial phenotype. In conclusion, [Zn(PipNONO)Cl] appears a promising therapeutic tool to control tumor growth and metastasis, by acting on both tumor and endothelial cells, reprogramming the cells toward their physiologic phenotypes.
Collapse
|
11
|
Nemeth DV, Baldini E, Sorrenti S, D’Andrea V, Bellini MI. Cancer Metabolism and Ischemia-Reperfusion Injury: Two Sides of the Same Coin. J Clin Med 2022; 11:jcm11175096. [PMID: 36079025 PMCID: PMC9457267 DOI: 10.3390/jcm11175096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/18/2022] [Accepted: 08/29/2022] [Indexed: 11/30/2022] Open
Abstract
Cancer cells are characterized by the reprogramming of certain cell metabolisms via activation of definite pathways and regulation of gene signaling. Ischemia-reperfusion injury (IRI) is characterized by tissue damage and death following a lack of perfusion and oxygenation. It is most commonly seen in the setting of organ transplantation. Interestingly, the microenvironments seen in cancer and ischemic tissues are quite similar, especially due to the hypoxic state that occurs in both. As a consequence, there is genetic signaling involved in response to IRI that has common pathways with cancer. Some of these changes are seen across the board with many cancer cells and are known as Hallmarks of Cancer, among which are aerobic glycolysis and the induction of angiogenesis. This literature review aims to compare the metabolic pathways that are altered in cancer tissues and in normal tissues subjected to IRI in order to find common adaptive processes and to identify key pathways that could represent a therapeutic target in both pathologies. By increasing our understanding of this relationship, clinical correlations can be made and applied practically to improve outcomes of transplanted organs, given the known association with acute rejection, delayed graft function, and poor graft survival. The following metabolic pathways are discussed in our review, both in the setting of cancer and IRI: apoptosis, glycolysis, and angiogenesis. The role of the immune system in both pathologies as well as mitochondrial function and the production of reactive oxygen species (ROS) are reviewed.
Collapse
Affiliation(s)
- Denise V. Nemeth
- School of Osteopathic Medicine, University of the Incarnate Word, San Antonio, TX 78235, USA
| | - Enke Baldini
- Department of Surgical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Salvatore Sorrenti
- Department of Surgical Sciences, Sapienza University of Rome, 00185 Rome, Italy
| | - Vito D’Andrea
- Department of Surgical Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (V.D.); (M.I.B.)
| | - Maria Irene Bellini
- Department of Surgical Sciences, Sapienza University of Rome, 00185 Rome, Italy
- Correspondence: (V.D.); (M.I.B.)
| |
Collapse
|
12
|
Salihi A, Al-Naqshabandi MA, Khudhur ZO, Housein Z, Hama HA, Abdullah RM, Hussen BM, Alkasalias T. Gasotransmitters in the tumor microenvironment: Impacts on cancer chemotherapy (Review). Mol Med Rep 2022; 26:233. [PMID: 35616143 PMCID: PMC9178674 DOI: 10.3892/mmr.2022.12749] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2021] [Accepted: 04/07/2022] [Indexed: 11/23/2022] Open
Abstract
Nitric oxide, carbon monoxide and hydrogen sulfide are three endogenous gasotransmitters that serve a role in regulating normal and pathological cellular activities. They can stimulate or inhibit cancer cell proliferation and invasion, as well as interfere with cancer cell responses to drug treatments. Understanding the molecular pathways governing the interactions between these gases and the tumor microenvironment can be utilized for the identification of a novel technique to disrupt cancer cell interactions and may contribute to the conception of effective and safe cancer therapy strategies. The present review discusses the effects of these gases in modulating the action of chemotherapies, as well as prospective pharmacological and therapeutic interfering approaches. A deeper knowledge of the mechanisms that underpin the cellular and pharmacological effects, as well as interactions, of each of the three gases could pave the way for therapeutic treatments and translational research.
Collapse
Affiliation(s)
- Abbas Salihi
- Department of Biology, College of Science, Salahaddin University-Erbil, Erbil, Kurdistan Region 44001, Iraq
- Center of Research and Strategic Studies, Lebanese French University, Erbil, Kurdistan Region 44002, Iraq
- Department of Microbiology, Tumor and Cell Biology (MTC), Biomedicum, Karolinska Institutet, SE-17165 Stockholm, Sweden
| | - Mohammed A. Al-Naqshabandi
- Department of Clinical Biochemistry, College of Health Sciences, Hawler Medical University, Erbil, Kurdistan Region 44001, Iraq
| | - Zhikal Omar Khudhur
- Department of Medical Analysis, Faculty of Applied Science, Tishk International University, Erbil, Kurdistan Region 44001, Iraq
| | - Zjwan Housein
- Department of Medical Laboratory Technology, Technical Health and Medical College, Erbil Polytechnique University, Erbil, Kurdistan Region 44002, Iraq
| | - Harmand A. Hama
- Department of Biology, Faculty of Education, Tishk International University, Erbil, Kurdistan Region 44002, Iraq
| | - Ramyar M. Abdullah
- College of Medicine, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region 44002, Iraq
| | - Twana Alkasalias
- General Directorate of Scientific Research Center, Salahaddin University-Erbil, Erbil, Kurdistan Region 44002, Iraq
- Department of Women's and Children's Health, Karolinska Institutet, Karolinska University Hospital, SE-17176 Stockholm, Sweden
| |
Collapse
|
13
|
Yu Z, Li M, Wang K, Gu Y, Guo S, Wang W, Ma Y, Liu H, Chen Y. Novel Hybrids of 3-Substituted Coumarin and Phenylsulfonylfuroxan as Potent Antitumor Agents with Collateral Sensitivity against MCF-7/ADR. J Med Chem 2022; 65:9328-9349. [PMID: 35737669 DOI: 10.1021/acs.jmedchem.2c00608] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Twenty-three new coumarin-furoxan hybrids were synthesized, which exhibited nanomole antiproliferation activities in A2780, A2780/CDDP, MCF-7/ADR, and MDA-MB-231. Among them, compound 9 showed the strongest collateral sensitivity to MCF-7/ADR with 499-fold potency compared with MCF-7. Notably, the solubility of compound 9 increased 70-fold compared with the lead 2. And preliminary pharmacological studies displayed that compound 9 obviously increased Rh123 accumulation in MCF-7/ADR and released NO to produce ROS in lysosomes, which were able to damage lysosomal membrane and induce apoptosis. These results reasonably explained that the collateral sensitivity of compound 9 to MCF-7/ADR was closely related to P-gp-mediated lysosome damage and apoptosis. Additionally, compound 9 showed a very weak cytotoxicity both in MCF-10A and hERG potassium channels and had a desirable safety in ion cyclotron resonance (ICR) mice. Hence, compound 9 was merited to further study for developing a desirable candidate against MDR MCF-7/ADR via a potential mechanism of collateral sensitivity in MDR cancer cell lines.
Collapse
Affiliation(s)
- Zhihui Yu
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Mengru Li
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ke Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China.,Pharmaceutical Sciences Division, School of Pharmacy,, University of Wisconsin-Madison, 777 Highland Avenue, Madison, Wisconsin 53705-2222, United States
| | - Yuting Gu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shiqi Guo
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Weijie Wang
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yulei Ma
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Hongrui Liu
- Department of Pharmacology, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Ying Chen
- Department of Medicinal Chemistry, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
14
|
Ouyang Q, Li Y, Mei S, Zhang Q, Li X, Luo H, Zhu Y, Wu K. Protective effects of GLHP from Gracilaria lemaneiformis against UVB-induced photodamage in human immortalized keratinocytes cells and BALB/c mice. Exp Gerontol 2021; 155:111550. [PMID: 34517082 DOI: 10.1016/j.exger.2021.111550] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 08/31/2021] [Accepted: 08/31/2021] [Indexed: 01/17/2023]
Abstract
Our previous study showed that the water-soluble heteropolysaccharide extracted from Gracilaria lemaneiformis (GLHP) has excellent anti-inflammation and anti-oxidant properties. This study explored the efficacy of GLHP against skin anti-photoaging in human immortalized keratinocytes (HaCaT) cells and BALB/c mice under UVB irradiation. Cell viability, antiapoptotic, reactive oxygen species (ROS) scavenging activity, mitochondrial membrane potential, and cell wound scratch assays were conducted, as well as assessment of inflammation markers and sun protection factors. The in vitro results showed that GLHP pretreatment significantly inhibited UVB-induced apoptosis, reversed the decrease of cell viability via downregulating the expression of apoptosis-related protein caspase-3, accelerated the migration of HaCaT cells, and promoted wound healing. Notably, the protective effect of GLHP may be associated with the scavenging of ROS and the decrease of mitochondrial membrane potential. Moreover, GLHP pretreatment significantly restrained the upregulation of iNOS (UVB-induced inflammation marker), suppressed the expression of P-ERK and NF-κB, and decreased the activity of MMPs, suggesting that it exerts the therapeutic effects by inhibiting the MAPK/NF-κB signal pathway. Results obtained after conducting the in vivo assay confirmed that GLHP could reverse the UVB-induced increase of epidermal thickness in BALB/c mice. In conclusion, this study shows that GLHP can be utilized as a safer resource in the manufacture of anti-aging cosmetics because it exerts excellent anti-photoaging effects.
Collapse
Affiliation(s)
- Qianqian Ouyang
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Yancai Li
- Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Si Mei
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Qian Zhang
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Xiaojun Li
- BoRui Saccharide Biotech Co. Ltd, Yangzhou 225000, PR China
| | - Hui Luo
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, PR China
| | - Yuzhen Zhu
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, PR China.
| | - Kefeng Wu
- Guangdong (Zhanjiang) Provincial Laboratory of Southern Marine Science and Engineering, 524023, PR China; Marine Biomedical Research Institution, Guangdong Medical University, Zhanjiang 524023, PR China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Guangdong Medical University, Zhanjiang 524023, PR China.
| |
Collapse
|
15
|
Minato A, Noguchi H, Ohnishi R, Tomisaki I, Nakayama T, Fujimoto N. Reduced Expression Level of GPX2 in T1 Bladder Cancer and its Role in Early-phase Invasion of Bladder Cancer. In Vivo 2021; 35:753-759. [PMID: 33622868 DOI: 10.21873/invivo.12316] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 11/10/2022]
Abstract
BACKGROUND/AIM The role of glutathione peroxidase 2 (GPX2) expression in urothelial carcinoma (UC) is rarely reported. The aim of this study was to assess the expression status of GPX2 in UC of the bladder. MATERIALS AND METHODS We collected samples from 112 patients treated with radical cystectomy for immunohistochemical study. RESULTS Following immunohistochemical analysis of the specimens, 86 (76.8%) had weak GPX2 expression. In cases with consistent GPX2 expression within the same lesion, the levels of GPX2 showed significant decreases from pTa to pT1 (47.1%) compared to those from pT1 to pT2 (5.9%) (p=0.017). Specimens obtained with transurethral resection before cancer progressed to muscle invasive bladder cancer showed that pT1 had a lower expression for GPX2 than that of pTa (63.3% vs. 93.3%; p=0.009). CONCLUSION The decrease in GPX2 expression among those with UC of the bladder may be involved in the early step of cancer invasion.
Collapse
Affiliation(s)
- Akinori Minato
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan;
| | - Hirotsugu Noguchi
- Department of Pathology, Field of Oncology Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Rei Ohnishi
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Ikko Tomisaki
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Toshiyuki Nakayama
- Department of Pathology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| | - Naohiro Fujimoto
- Department of Urology, School of Medicine, University of Occupational and Environmental Health, Kitakyushu, Japan
| |
Collapse
|
16
|
Xie J, Chen L, Huang D, Yue W, Chen J, Liu C. A nitric oxide-releasing prodrug promotes apoptosis in human renal carcinoma cells: Involvement of reactive oxygen species. OPEN CHEM 2021. [DOI: 10.1515/chem-2020-0075] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Abstract
Background
JS-K is a nitric oxide (NO)-releasing prodrug of the O2-arylated diazeniumdiolate group that shows pronounced cytotoxicity and antitumor properties in numerous cancer models, including in vitro as well as in vivo. Reactive oxygen species (ROS) induce carcinogenesis by altering the redox status, causing increment in vulnerability to oxidative stress.
Material and methods
To determine the effect of JS-K, a glutathione S-transferase (GST)-activated NO-donor prodrug, on the induction of ROS accumulation, proliferation, and apoptosis in human renal carcinoma cells, we measured the changes of cell proliferation, apoptosis, ROS growth, and initiation of the mitochondrial signaling pathway before and after JS-K treatment.
Results
In vitro, dose- and time-dependent development of renal carcinoma cells were controlled for JS-K, and JS-K also triggered ROS aggregation and cell apoptosis. Treatment with JS-K induces the levels of pro-apoptotic proteins (Bak and Bax) and decrease the number of anti-apoptotic protein (Bcl-2). In fact, JS-K-induced apoptosis was reversed by the antioxidant N-acetylcysteine, and oxidized glutathione, a pro-oxidant, improved JS-K-induced apoptosis. Finally, we demonstrated that in renal carcinoma cells, JS-K improved the chemosensitivity of doxorubicin.
Conclusion
Our data indicate that JS-K-released NO induce apoptosis of renal carcinoma cells by increasing ROS levels.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Urology, Zhujiang Hospital, Southern Medical University , No. 253, Industrial Road , Guangzhou , 510282, Guangdong , China
| | - Lieqian Chen
- Department of Urology, The First Hospital of Huizhou , No. 20, Sanxin Road , Huizhou , 516000, Guangdong , China
| | - Dongqiang Huang
- Department of Urology, The First Hospital of Huizhou , No. 20, Sanxin Road , Huizhou , 516000, Guangdong , China
| | - Weiwei Yue
- Department of Urology, The First Hospital of Huizhou , No. 20, Sanxin Road , Huizhou , 516000, Guangdong , China
| | - Jingyu Chen
- Department of Urology, The First Hospital of Huizhou , No. 20, Sanxin Road , Huizhou , 516000, Guangdong , China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University , No. 253, Industrial Road , Guangzhou , 510282, Guangdong , China
| |
Collapse
|
17
|
Chen C, Guo Z, Ma G, Ma J, Zhang Z, Yu Q, Han L. Lysosomal Fe2+contributes to myofibrillar protein degradation through mitochondrial-dysfunction-induced apoptosis. Lebensm Wiss Technol 2021. [DOI: 10.1016/j.lwt.2021.111197] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
|
18
|
Betulinic Acid Restricts Human Bladder Cancer Cell Proliferation In Vitro by Inducing Caspase-Dependent Cell Death and Cell Cycle Arrest, and Decreasing Metastatic Potential. Molecules 2021; 26:molecules26051381. [PMID: 33806566 PMCID: PMC7961550 DOI: 10.3390/molecules26051381] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 02/26/2021] [Accepted: 02/27/2021] [Indexed: 12/15/2022] Open
Abstract
Betulinic acid (BA) is a naturally occurring pentacyclic triterpenoid and generally found in the bark of birch trees (Betula sp.). Although several studies have been reported that BA has diverse biological activities, including anti-tumor effects, the underlying anti-cancer mechanism in bladder cancer cells is still lacking. Therefore, this study aims to investigate the anti-proliferative effect of BA in human bladder cancer cell lines T-24, UMUC-3, and 5637, and identify the underlying mechanism. Our results showed that BA induced cell death in bladder cancer cells and that are accompanied by apoptosis, necrosis, and cell cycle arrest. Furthermore, BA decreased the expression of cell cycle regulators, such as cyclin B1, cyclin A, cyclin-dependent kinase (Cdk) 2, cell division cycle (Cdc) 2, and Cdc25c. In addition, BA-induced apoptosis was associated with mitochondrial dysfunction that is caused by loss of mitochondrial membrane potential, which led to the activation of mitochondrial-mediated intrinsic pathway. BA up-regulated the expression of Bcl-2-accociated X protein (Bax) and cleaved poly-ADP ribose polymerase (PARP), and subsequently activated caspase-3, -8, and -9. However, pre-treatment of pan-caspase inhibitor markedly suppressed BA-induced apoptosis. Meanwhile, BA did not affect the levels of intracellular reactive oxygen species (ROS), indicating BA-mediated apoptosis was ROS-independent. Furthermore, we found that BA suppressed the wound healing and invasion ability, and decreased the expression of Snail and Slug in T24 and 5637 cells, and matrix metalloproteinase (MMP)-9 in UMUC-3 cells. Taken together, this is the first study showing that BA suppresses the proliferation of human bladder cancer cells, which is due to induction of apoptosis, necrosis, and cell cycle arrest, and decrease of migration and invasion. Furthermore, BA-induced apoptosis is regulated by caspase-dependent and ROS-independent pathways, and these results provide the underlying anti-proliferative molecular mechanism of BA in human bladder cancer cells.
Collapse
|
19
|
Zhao Y, Li Z, Tang H, Lin S, Zeng W, Ye D, Zeng X, Luo Q, Li J, Ao Z, Mo J, Chen L, Yang Y, Huang Y, Liu J. [Mn(PaPy2Q)(NO)]ClO 4, a Near-Infrared Light activated release of Nitric Oxide drug as a nitric oxide donor for therapy of human prostate cancer cells in vitro and in vivo. Biomed Pharmacother 2021; 137:111388. [PMID: 33761607 DOI: 10.1016/j.biopha.2021.111388] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/31/2021] [Accepted: 02/09/2021] [Indexed: 12/20/2022] Open
Abstract
This study was the first to investigate the synthesis of near-infrared light-sensitive NO prodrug [Mn(PaPy2Q)(NO)]ClO4, and detection the amount of NO released by the drug in different time and near infrared light (10 mW, 20 mW). It showed that with the increase of light power, the time required for the drug to release NO was shortened, and we selected 20 mW, 10 min as a follow-up study of light power and irradiation time while ensuring the near-infrared light did not affect tumor cells. The cells were irradiated with 20 mW of near-infrared light for 10 min at 6 h after treatment with the drug on PC-3, LNCaP and 22RV1 cells, and NO concentration and cell survival rate were tested at 12 h, 24 h and 48 h. Experiments showed that NO concentration remained stable within 48 h and [Mn(PaPy2Q)(NO)]ClO4 inhibited the proliferation of cells in a concentration and time-dependent manner. Then we also found that [Mn(PaPy2Q)(NO)]ClO4 increased the expression of apoptosis-related proteins (PARP, Bax, Caspase 3/9), inhibited the expression of BCl-2 and increased the activity level of Caspase 3/7, which showed [Mn(PaPy2Q)(NO)]ClO4 promoted prostate cancer cells apoptosis. Next, the results in xenograft mouse model showed that [Mn(PaPy2Q)(NO)]ClO4 also had anti-prostate cancer effects in vivo, and the NO concentration increased in the tumor after near-infrared light irradiation. After [Mn(PaPy2Q)(NO)]ClO4 treatment 6 weeks, tumor volume was significantly reduced, Ki67 and BrdU protein expression was significantly reduced. TUNEL assay results showed that [Mn(PaPy2Q)(NO)]ClO4 could promote the apoptosis of solid tumors in vivo and in a concentration-dependent manner.
Collapse
Affiliation(s)
- Yuwan Zhao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhuo Li
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Huancheng Tang
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Shanhong Lin
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Wenfeng Zeng
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Dongcai Ye
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Xin Zeng
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Qiuming Luo
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jianwei Li
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zhixian Ao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Jierong Mo
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Lixin Chen
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yiqiu Yang
- School of Pharmacy, Guangdong Medical University, Xincheng Ave, Songshan Lake Technology Park, Dongguan 523808, China
| | - Yunsheng Huang
- School of Pharmacy, Guangdong Medical University, Xincheng Ave, Songshan Lake Technology Park, Dongguan 523808, China.
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| |
Collapse
|
20
|
Qiu M, Liu J, Su Y, Liu J, Wu C, Zhao B. Aloperine Induces Apoptosis by a Reactive Oxygen Species Activation Mechanism in Human Ovarian Cancer Cells. Protein Pept Lett 2021; 27:860-869. [PMID: 32196436 DOI: 10.2174/0929866527666200320094313] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2019] [Revised: 02/01/2020] [Accepted: 02/01/2020] [Indexed: 12/28/2022]
Abstract
BACKGROUND Ovarian cancer is the most lethal gynecologic malignancy worldwide with poor prognosis owing to chemotherapy resistance and cancer relapse. Hence, there is an urgent need to develop novel anticancer agents against ovarian cancer. OBJECTIVE The aim of this research is to investigate the possible anticancer activity of aloperine, an active ingredient from a traditional Chinese medicine Sophora alopecuroides, and to explore the possible Reactive Oxygen Species (ROS)-related mechanism. METHODS Cell viability, cytotoxicity, apoptosis, ROS generation, and oxidant stress indicators were analyzed. RESULTS Our results demonstrated that aloperine significantly induced inhibition of cell viability, promoted cytotoxicity and mitochondrial-related apoptosis, and increased ROS generation in ovarian cancer cells. Furthermore, the antioxidant α-lipoic acid reversed apoptosis in aloperinetreated cells. In addition, we identified hydrogen peroxide as the main type of ROS, and the antioxidant catalase suppressed the apoptotic inducing effect of aloperine whereas hydrogen peroxide supplement exacerbated the effect of aloperine in ovarian cancer cells. CONCLUSION Taken together, our results indicated that aloperine could exert anti-ovarian cancer cell activity through a reactive oxygen species activation mechanism and suggested aloperine as a potential agent against ovarian cancer.
Collapse
Affiliation(s)
- Mingning Qiu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Laboratory of Urology, Guangdong Medical University, Zhanjiang, China
| | - Jie Liu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China.,Laboratory of Urology, Guangdong Medical University, Zhanjiang, China
| | - Yongxia Su
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, China
| | - Chenchen Wu
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
21
|
Dietary isothiocyanates inhibit cancer progression by modulation of epigenome. Semin Cancer Biol 2021; 83:353-376. [PMID: 33434642 DOI: 10.1016/j.semcancer.2020.12.021] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 12/13/2020] [Accepted: 12/27/2020] [Indexed: 12/15/2022]
Abstract
Cell cycle, growth, survival and metabolism are tightly regulated together and failure in cellular regulation leads to carcinogenesis. Several signaling pathways like the PI3K, WNT, MAPK and NFKb pathway exhibit aberrations in cancer and help achieve hallmark capabilities. Clinical research and in vitro studies have highlighted the role of epigenetic alterations in cancer onset and development. Altered gene expression patterns enabled by changes in DNA methylation, histone modifications and RNA processing have proven roles in cancer hallmark acquisition. The reversible nature of epigenetic processes offers robust therapeutic targets. Dietary bioactive compounds offer a vast compendium of effective therapeutic moieties. Isothiocyanates (ITCs) sourced from cruciferous vegetables demonstrate anti-proliferative, pro-apoptotic, anti-inflammatory, anti-migratory and anti-angiogenic effect against several cancers. ITCs also modulate the redox environment, modulate signaling pathways including PI3K, MAPK, WNT, and NFkB. They also modulate the epigenetic machinery by regulating the expression and activity of DNA methyltransferases, histone modifiers and miRNA. This further enhances their transcriptional modulation of key cellular regulators. In this review, we comprehensively assess the impact of ITCs such as sulforaphane, phenethyl isothiocyanate, benzyl isothiocyanate and allyl isothiocyanate on cancer and document their effect on various molecular targets. Overall, this will facilitate consolidation of the current understanding of the anti-cancer and epigenetic modulatory potential of these compounds and recognize the gaps in literature. Further, we discuss avenues of future research to develop these compounds as potential therapeutic entities.
Collapse
|
22
|
Liu L, Xing Y, Cao M, Xu J, Chen J. Exogenous NO induces apoptosis of hepatocellular carcinoma cells via positive p38/JNK signaling pathway and negative ERK signaling pathways. Mol Cell Biochem 2021; 476:1651-1661. [PMID: 33420899 DOI: 10.1007/s11010-020-04032-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 12/22/2020] [Indexed: 12/24/2022]
Abstract
JS-K as an exogenous NO donor could release NO after activation by glutathione S-transferases (GSTs). The present study explores the effects of JS-K on MAPK pathway in HepG2 and Bel-7402 cells. JS-K significantly prompted apoptosis and SB203580 (a p38 inhibitor) and SP600125 (a JNK inhibitor) prior to JS-K could partly reverse apoptosis and activation of cleaved-caspase-3 and cleaved PARP. However, U0126 (a MEK inhibitor) strengthened the cell apoptosis and the expressions of cleaved-caspase-3 and cleaved PARP. JS-K caused phosphorylation of p38 MAPK and JNK but attenuated phosphorylation of ERK, which were reversed by Carboxy-PTIO (a NO scavenger). Meanwhile, the phosphorylation of HSP27, c-JUN and ATF-2 were activated in JS-K-treated cells. SB203580 and SP600125 could attenuate phosphorylation of p38 MAPK and JNK, respectively. The phosphorylation in downstream substrates of p38 MAPK and JNK was also abolished by SB203580 and SP600125 in JS-K-treated cells. Additionally, JS-K decreased phosphorylation of c-Raf, which subsequently caused a decrease of MEK1/2 phosphorylation. Several downstream targets of ERK1/2 including p90RSK and transcription factors (e.g., Elk-1, c-Myc and c-Fos) were inhibited. U0126 potentiated JS-K-induced inhibitory effect of Raf/MEK/ERK pathway. The same results were also observed in the downstream substrates of ERK1/2 including p90RSK, Elk-1, c-Myc and c-Fos. Moreover, Carboxy-PTIO abolished the inhibitory effect of Raf/MEK/ERK pathway triggered by JS-K. Finally, JS-K significantly suppressed the growth of rat primary hepatic carcinoma via MAPK pathway in vivo. Taken together, JS-K can induce hepatocellular carcinoma cells apoptosis through its activation of JNK and p38 MAPK and inactivation of Raf/MEK/ERK signaling pathways.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China.
| | - Yihao Xing
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Mengyao Cao
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Jinglei Xu
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| | - Jingjing Chen
- Department of Pharmacy, School of Basic Medical Sciences, Henan University of Science and Technology, 263 Kaiyuan Avenue, Luoyang, 471023, China
| |
Collapse
|
23
|
Suzuki S, Cohen SM, Arnold LL, Pennington KL, Gi M, Kato H, Naiki T, Naiki-Ito A, Wanibuchi H, Takahashi S. Cell proliferation of rat bladder urothelium induced by nicotine is suppressed by the NADPH oxidase inhibitor, apocynin. Toxicol Lett 2021; 336:32-38. [PMID: 33176187 DOI: 10.1016/j.toxlet.2020.11.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 10/19/2020] [Accepted: 11/04/2020] [Indexed: 11/27/2022]
Abstract
Tobacco smoking is a major risk factor for human cancers including urinary bladder carcinoma. In a previous study, nicotine enhanced rat urinary bladder carcinogenesis in a two-stage carcinogenesis model. Nicotine also induced cytotoxicity in the bladder urothelium in a short-term study. In the present study, male rats were treated with nicotine (40 ppm) in drinking water co-administered with the NADPH oxidase inhibitor, apocynin (0, 250 or 750 mg/kg) in diet for 4 weeks. The apocynin treatment induced no clinical toxic effects. Reduction of reactive oxygen species (ROS) by apocynin was confirmed by immunohistochemistry of 8-OHdG in the bladder urothelium. Incidences of simple hyperplasia, cell proliferation and apoptosis were reduced by apocynin treatment in the bladder urothelium. However, despite reduction of cell proliferation (labeling index), apocynin did not affect the incidence of simple hyperplasia, apoptosis, or ROS generation in the kidney pelvis urothelium, in addition to 8-OHdG positivity induced by nicotine being lower. In vitro, apocynin (500 μM) reduced ROS generation, but induced cell proliferation in bladder cancer cell lines (T24 and UMUC3 cells). These data suggest that oxidative stress may play a role in the cell proliferation of the bladder urothelium induced by nicotine.
Collapse
Affiliation(s)
- Shugo Suzuki
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan; Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan.
| | - Samuel M Cohen
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-5900, USA; Havlik-Wall Professor of Oncology, University of Nebraska Medical Center, Omaha, NE, 68198-3135, USA
| | - Lora L Arnold
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, 68198-5900, USA
| | - Karen L Pennington
- Department of Pharmaceutical Sciences, University of Nebraska Medical Center, Omaha, NE, 68198- 6849, USA
| | - Min Gi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| | - Hideki Wanibuchi
- Department of Molecular Pathology, Osaka City University Graduate School of Medicine, Osaka, Osaka 545-8585, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University Graduate School of Medical Sciences, Nagoya, Aichi 467-8601, Japan
| |
Collapse
|
24
|
Cinnamaldehyde induces endogenous apoptosis of the prostate cancer-associated fibroblasts via interfering the Glutathione-associated mitochondria function. Med Oncol 2020; 37:91. [DOI: 10.1007/s12032-020-01417-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 09/04/2020] [Indexed: 01/03/2023]
|
25
|
Liu J, Zhao Y, Xia J, Qiu M. Matrine induces toxicity in mouse liver cells through an ROS-dependent mechanism. Res Vet Sci 2020; 132:308-311. [PMID: 32717422 DOI: 10.1016/j.rvsc.2020.07.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 07/05/2020] [Accepted: 07/10/2020] [Indexed: 02/07/2023]
Abstract
Matrine is major active component in Sophora flavescens Ait that plays pharmacological activities against inflammation, tumors and virus. However, potential toxicity of matrine and its possible toxic mechanisms have not been carefully studied. The aim of the study is to assess the toxicity of matrine on mouse liver cells and to investigate the potential ROS-associated mechanisms. Mice were randomly divided into three groups: vehicle control (normal saline), low-dose (50 mg/kg), and high dose (100 mg/kg) groups. Mice in each group were intraperitoneally injected with matrine daily for 7 d. The livers were collected for analysis of histopathological changes and HO-1 protein expression. Serum was collected for analysis of aspartate aminotransferase and alanine aminotransferase activities. Mouse liver NCTC cells were treated with matrine for certain time, and cell viability, cytotoxicity, apoptosis, expression of proteins, activities of caspase-3 and caspase-9, and levels of ROS generation, mitochondrial membrane potential, and ATP were examined. Increased activities of AST and ALT in serum, and vacuolar degeneration of cytoplasm in liver tissues were observed after treatment. Suppression of cell viability, increase of cytotoxicity, induction of apoptosis, alteration in the expression of apoptotic-related proteins, and activation of caspase-3 and caspase-9 were shown in matrine-treated NCTC cells. Furthermore, matrine induced ROS generation, and suppressed mitochondrial membrane potential and ATP levels, however, the antioxidant N-acetylcysteine reversed matrine-induced hepatotoxicity and ROS generation. These findings suggested that matrine stimulated the generation of ROS, which was possibly involved in matrine-induced toxicity in mouse liver cells in vitro and in vivo.
Collapse
Affiliation(s)
- Jie Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, 524001, China
| | - Yuwan Zhao
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, 524001, China
| | - Juan Xia
- Laboratory of Hematologic Diseases, Guangdong Medical University, Zhanjiang, 524001, China
| | - Mingning Qiu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, 524001, China.
| |
Collapse
|
26
|
Liu H, Xiong X, Zhu T, Zhu Y, Peng Y, Zhu X, Wang J, Chen H, Chen Y, Guo A. Differential nitric oxide induced by Mycobacterium bovis and BCG leading to dendritic cells apoptosis in a caspase dependent manner. Microb Pathog 2020; 149:104303. [PMID: 32504845 DOI: 10.1016/j.micpath.2020.104303] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 03/14/2020] [Accepted: 05/29/2020] [Indexed: 12/25/2022]
Abstract
Dendritic cells (DCs) are critical for both innate and adaptive immunity. Meanwhile, nitric oxide (NO) is a member of reactive nitrogen species (RNS) generally considered to play a key role in the bactericidal process in innate immunity against Mycobacterium tuberculosis complex infection. The present study therefore investigated the mechanism of NO production in murine DCs induced by Mycobacterium bovis (M.bovis) and its attenuated strain Bacillus Calmette-Guérin (BCG) infection. The expression of genes Slc7A1, Slc7A2, iNOS, and ArgI essential to NO synthesis was up-regulated in M.bovis/BCG infected DCs. IFN-γ addition further increased, while the iNOS inhibitor L-NMMA significantly inhibited their expression. Accordingly, the end products of arginine metabolism, NO and urea, were found to be significantly increased. In addition, BCG induced significantly higher levels of apoptosis in DCs compared to M.bovis shown by higher levels of DNA fragmentation using flow cytometry and release of mitochondrial Cytochrome C, and up-regulation of the genes caspase-3, caspase-8, caspase-9 and dffa critical to apoptosis by qRT-PCR detection and western blot analysis. Furthermore, IFN-γ increased, but L-NMMA decreased apoptosis of M.bovis/BCG infected DCs. In addition, mycobacterial intracellular survival was significantly reduced by IFN-γ treatment in BCG infected DCs, while slightly increased by L-NMMA treatment. Taken altogether, our data show that NO synthesis was differentially increased and associated with apoptosis in M.bovis/BCG infected DCs. These findings may significantly contribute to elucidate the pathogenesis of M.bovis.
Collapse
Affiliation(s)
- Han Liu
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Xuekai Xiong
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Tingting Zhu
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Yifan Zhu
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Yongchong Peng
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Xiaojie Zhu
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Jieru Wang
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Huanchun Chen
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Yingyu Chen
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China.
| | - Aizhen Guo
- The National Key Laboratory of Agricultural Microbiology, Wuhan, China; College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, China; Hubei International Scientific and Technological Cooperation Base of Veterinary Epidemiology, Huazhong Agricultural University, Wuhan, 430070, China; Key Laboratory of Development of Veterinary Diagnostic Products, Key Laboratory of Ruminant Bio-products, Huazhong Agricultural University, Wuhan, 430070, China; Ministry of Agriculture and Rural Affairs, Huazhong Agricultural University, Wuhan, 430070, China.
| |
Collapse
|
27
|
Zhao Y, Luo Q, Mo J, Li J, Ye D, Ao Z, Chen L, Liu J. Metformin in combination with JS-K inhibits growth of renal cell carcinoma cells via reactive oxygen species activation and inducing DNA breaks. J Cancer 2020; 11:3701-3712. [PMID: 32328174 PMCID: PMC7171495 DOI: 10.7150/jca.36372] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2019] [Accepted: 02/06/2020] [Indexed: 02/06/2023] Open
Abstract
Metformin (MET) is taken as a principal medication for remedying Type 2 diabetes mellitus. Its anti-tumor effect has been reported increasingly, but the precise mechanism of it remains unclear. This study aims to explore the efficacy of MET and MET combined with nitric oxide donor prodrug JS-K on the proliferation, apoptosis, and DNA damage in human renal cell carcinoma (RCC) cells, and investigate the possible molecular mechanism involved. The cell proliferation was tested through methyl-tetrazolium assay and cell apoptosis was ascertained by flow cytometry. The dihydroethidium and JC-1 fluorescent methods were used to detect Reactive oxygen species (ROS) and mitochondrial transmembrane potential (Δψm), respectively. Proteins associated with apoptosis and DNA damage were evaluated by Western blotting. Results showed that MET and JS-K could suppress cell growth, and the inhibition concentration 50 of treatment with MET combined with JS-K (MET + JS-K) showed more toxicity than individual agents on RCC cells. This augmented toxicity was associated with intracellular reactive oxygen species (ROS) level, mitochondrial membrane potential alteration, and induced DNA breaks. The results of Western blotting showed that the expression level of pro-apoptotic proteins, such as Bax, Bak, caspase-3, and caspase-9, was up-regulated, and the anti-apoptotic protein Bcl-2 was down-regulated after treatment using MET alone and MET + JS-K, correspondingly. Moreover, MET + JS-K inhibited the expression of cellular PCNA and Rad51, and immunofluorescence analysis of γH2AX proved that MET + JS-K enhanced DNA damage. In summary, the results of this research indicated that MET and JS-K inhibited RCC cell growth by activating ROS, targeting mitochondria-dependent apoptotic pathways, and inducing DNA breaks.
Collapse
Affiliation(s)
- Yuwan Zhao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Qiuming Luo
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Jierong Mo
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Jianwei Li
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Dongcai Ye
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Zhixian Ao
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Lixin Chen
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| | - Jianjun Liu
- Laboratory of Urology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, Guangdong 524001, China
| |
Collapse
|
28
|
Bonavida B. Sensitizing activities of nitric oxide donors for cancer resistance to anticancer therapeutic drugs. Biochem Pharmacol 2020; 176:113913. [PMID: 32173364 DOI: 10.1016/j.bcp.2020.113913] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2019] [Accepted: 03/10/2020] [Indexed: 02/08/2023]
Abstract
Cancer is not a single disease but it constitutes a large variety of different types that are also different from each other phenotypically and molecularly. Although the standard treatments have resulted in clinical responses in a subset of patients, though, many patients relapse and no longer respond to further treatments. Hence, both the innate and adaptive resistance to treatments are the main challenges in today's treatment strategies. Noteworthy, several novel treatment strategies, particularly immunotherapies, used alone or in combination, have been developed and that have significantly improved the therapeutic response of many unresponsive cancer patients. Nevertheless, even with the latest new developments of therapeutics that were effective in a larger subset of patients, there is still an urgent need to treat the remaining unresponsive subset of patients. This requires the development of new targeting agents of superior antitumor activities that will lead to overcoming the unaffected resistance by current treatments. There has been accumulating evidence suggesting nitric oxide donors as such targeting agents and considering their pleiotropic antitumor activities, including both the reversal of chemo and immuno-resistance of various unresponsive resistant cancers. The in vitro and in vivo preclinical findings corroborate the sensitizing antitumor activities of nitric oxide donors. In addition, a few clinical findings with NO donors that have been applied in patients have corroborated their antitumor and sensitizing activities in combination with standard therapies. In this review, the role and underlying mechanisms by which nitric oxide donors sensitize cancer resistant cells to both chemotherapy and immunotherapy are briefly described.
Collapse
Affiliation(s)
- Benjamin Bonavida
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
29
|
Wu W, Chen M, Luo T, Fan Y, Zhang J, Zhang Y, Zhang Q, Sapin-Minet A, Gaucher C, Xia X. ROS and GSH-responsive S-nitrosoglutathione functionalized polymeric nanoparticles to overcome multidrug resistance in cancer. Acta Biomater 2020; 103:259-271. [PMID: 31846803 DOI: 10.1016/j.actbio.2019.12.016] [Citation(s) in RCA: 73] [Impact Index Per Article: 14.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2019] [Revised: 12/11/2019] [Accepted: 12/12/2019] [Indexed: 12/27/2022]
Abstract
Multidrug resistance of cancer cells is one of the major obstacle for chemotherapeutic efficiency. Nitric oxide (NO) has raised the potential to overcome multidrug resistance (MDR) with low side effects. Herein, we report a reactive oxygen species (ROS) and glutathione (GSH) responsive nanoparticle for the delivery of NO prodrug such as S-nitrosoglutathione (GSNO), which was chemically conjugated to an amphiphilic block copolymer. The GSNO functionalized nanoparticles show high NO loading capacity, good stability and sustained NO release with specific GSH activated NO-releasing kinetics. Such GSNO functionalized nanoparticles delivered doxorubicin (DOX) in a ROS triggered manner and increased the intracellular accumulation of DOX. However, in normal healthy cells, showing physiological concentrations of ROS, these nanoparticles presented good biocompatibility. The present work indicated that these multifunctional nanoparticles can serve as effective co-delivery platforms of NO and DOX to selectively kill chemo-resistant cancer cells through increasing chemo-sensitivity. STATEMENT OF SIGNIFICANCE: In this work, we constructed nitric oxide donor (S-nitrosoglutathione, GSNO) functionalized amphiphilic copolymer (PEG-PPS-GSNO) to deliver doxorubicin (DOX). The developed PEG-PPS-GSNO@DOX nanoparticles presented high NO capacity, ROS triggered DOX release and GSH triggered NO release. Thus NO reversed the chemo-resistance in HepG2/ADR cells increasing intrcellular accumulation of DOX. Furthermore, these PEG-PPS-GSNO@DOX nanoparticles exhibited biocompatibility to healthy cells and toxicity to cancer cells, due to elevated ROS.
Collapse
|
30
|
Wu K, Lu Y, Mei S, Wang P, Ouyang P, Liao X, Ye H, Ma X. Protective effects of Gracilaria lemaneiformis extract against ultraviolet B-induced damage in HaCaT cells. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_437_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
31
|
Hays E, Bonavida B. Nitric Oxide-Mediated Enhancement and Reversal of Resistance of Anticancer Therapies. Antioxidants (Basel) 2019; 8:E407. [PMID: 31533363 PMCID: PMC6769868 DOI: 10.3390/antiox8090407] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Revised: 09/11/2019] [Accepted: 09/13/2019] [Indexed: 12/13/2022] Open
Abstract
In the last decade, immune therapies against human cancers have emerged as a very effective therapeutic strategy in the treatment of various cancers, some of which are resistant to current therapies. Although the clinical responses achieved with many therapeutic strategies were significant in a subset of patients, another subset remained unresponsive initially, or became resistant to further therapies. Hence, there is a need to develop novel approaches to treat those unresponsive patients. Several investigations have been reported to explain the underlying mechanisms of immune resistance, including the anti-proliferative and anti-apoptotic pathways and, in addition, the increased expression of the transcription factor Yin-Yang 1 (YY1) and the programmed death ligand 1 (PD-L1). We have reported that YY1 leads to immune resistance through increasing HIF-1α accumulation and PD-L1 expression. These mechanisms inhibit the ability of the cytotoxic T-lymphocytes to mediate their cytotoxic functions via the inhibitory signal delivered by the PD-L1 on tumor cells to the PD-1 receptor on cytotoxic T-cells. Thus, means to override these resistance mechanisms are needed to sensitize the tumor cells to both cell killing and inhibition of tumor progression. Treatment with nitric oxide (NO) donors has been shown to sensitize many types of tumors to chemotherapy, immunotherapy, and radiotherapy. Treatment of cancer cell lines with NO donors has resulted in the inhibition of cancer cell activities via, in part, the inhibition of YY1 and PD-L1. The NO-mediated inhibition of YY1 was the result of both the inhibition of the upstream NF-κB pathway as well as the S-nitrosylation of YY1, leading to both the downregulation of YY1 expression as well as the inhibition of YY1-DNA binding activity, respectively. Also, treatment with NO donors induced the inhibition of YY1 and resulted in the inhibition of PD-L1 expression. Based on the above findings, we propose that treatment of tumor cells with the combination of NO donors, at optimal noncytotoxic doses, and anti-tumor cytotoxic effector cells or other conventional therapies will result in a synergistic anticancer activity and tumor regression.
Collapse
Affiliation(s)
- Emily Hays
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095, USA.
| | - Benjamin Bonavida
- Department of Microbiology, Immunology and Molecular Genetics, David Geffen School of Medicine, University of California Los Angeles, CA 90095, USA.
| |
Collapse
|
32
|
Rasheduzzaman M, Yin H, Park SY. Cardiac glycoside sensitized hepatocellular carcinoma cells to TRAIL via ROS generation, p38MAPK, mitochondrial transition, and autophagy mediation. Mol Carcinog 2019; 58:2040-2051. [PMID: 31392779 DOI: 10.1002/mc.23096] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2019] [Revised: 07/17/2019] [Accepted: 07/26/2019] [Indexed: 12/18/2022]
Abstract
A major concern in the clinical application of tumor necrosis factor related apoptosis-inducing ligand (TRAIL) in tumors is the development of resistance. Therefore, agents that can potentially restore TRAIL sensitivity are important therapeutic targets for cancer treatment. Herein, we evaluated lanatoside c and digoxin, both of which are widely used cardiac glycosides (CGs), for their ability to sensitize human hepatocellular carcinoma cells (Huh-7 and HepG2) through TRAIL-induced apoptosis. CGs functionalize TRAIL as shown by its effect on intracellular reactive oxygen species (ROS) generation, which damages mitochondrial integrity and thereby confers intrinsic apoptotic caspase cascade during combined treatment. Caspase activation is dependent on ROS as shown by the ability of CGs to generate ROS and the ROS-N-acetylcysteine (NAC) relationship, which inhibits apoptosis during cotreatment by preventing the formation of caspase-8 and -3. Furthermore, CGs triggered p38MAPK phosphorylation and NAC pre-exposure blocked p38MAPK phosphorylation, which demonstrated that p38MAPK was dependent upon ROS generation. Additionally, CGs were found to be potent inducers of AMPK-mediated protective autophagy as pharmacological and genetic autophagy inhibition reached the higher threshold of TRAIL-mediated apoptosis. Finally, CGs downregulated the expression of the antiapoptotic protein Bcl-2 and increased the translocation of proapoptotic protein cytochrome c, thereby inducing apoptosis. Collectively, these results indicate that CGs potentiate the enhanced cytotoxic capacity to TRAIL through ROS generation, p38MAPK phosphorylation, cell survival protein downregulation, and protective autophagy inhibition.
Collapse
Affiliation(s)
- Mohammad Rasheduzzaman
- School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, Brisbane, Queensland, Australia.,Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Honghua Yin
- Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| | - Sang-Youel Park
- Biosafety Research Institute, Department of Veterinary Medicine, College of Veterinary Medicine, Chonbuk National University, Iksan, Jeonbuk, South Korea
| |
Collapse
|
33
|
Liu B, Huang X, Li Y, Liao W, Li M, Liu Y, He R, Feng D, Zhu R, Kurihara H. JS-K, a nitric oxide donor, induces autophagy as a complementary mechanism inhibiting ovarian cancer. BMC Cancer 2019; 19:645. [PMID: 31262254 PMCID: PMC6604176 DOI: 10.1186/s12885-019-5619-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 04/16/2019] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the second most frequent gynecological cancer and is associated with a poor prognosis because OC progression is often asymptoma-tic and is detected at a late stage. There remains an urgent need for novel targeted therapies to improve clinical outcomes in ovarian cancer. As a nitric oxide prodrug, JS-K is reported highly cytotoxic to human cancer cells such as acute myeloid leukemia, multiple myeloma and breast cancer. This study is aim to investigate the influence of JS-K on proliferation and apoptosis in ovarian cancer cells and explored possible autophagy-related mechanisms, which will contribute to future ovarian cancer therapy and supply theory support that JS-K holds great promise as a novel therapeutic agent against ovarian cancer. METHODS The cytotoxicity, extracellular ROS/RNS activity and apoptotic effect of JS-K and indicated inhibitors on ovarian cancer cells in vitro were evaluated by MTT assay, extracellular ROS/RNS assay, caspases activities assay and western blot. Further autophagy effect of JS-K and indicated inhibitors were examined by MTT assay, cell transfection, immunofluorescence analysis, transmission electron microscopy (TEM) analysis and western blot on ovarian cancer cells in vitro. In vivo, the BALB/c-nude female mice with SKOV3 ovarian cancer cells xenograft were used to examine the efficacy of JS-K treatment on tumor growth. PCNA and p62 proteins were analyzed by immunohistochemistry. RESULTS In vitro, JS-K inhibited the proliferation of ovarian cancer cells, induced apoptosis and cell nucleus shrinkage, enhanced the enzymatic activity of caspase-3/7/8/9, and significantly increased the production of ROS/RNS in ovarian cancer A2780 and SKOV3 cells, these effects were attenuated by inhibition of NAC. In addition, JS-K induced autophagy-related proteins and autophagosomes changes in ovarian cancer A2780 and SKOV3 cells. In vivo, JS-K inhibited tumor growth, decreased p62 protein expression and increased the expression levels of PCNA in xenograft models which were established using SKOV3 ovarian cancer cells. CONCLUSION Taken together, we demonstrated that ROS/RNS stress-mediated apoptosis and autophagy are mechanisms by which SKOV3 cells undergo cell death after treatment with JS-K in vitro. Moreover, JS-K inhibited SKOV3 tumor growth in vivo. An alternative therapeutic approach for triggering cell death in cancer cells could constitute a useful multimodal therapies for treating ovarian cancer, which is known for its resistance to apoptosis-inducing drugs.
Collapse
Affiliation(s)
- Bin Liu
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Xiaojie Huang
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Yifang Li
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| | - Weiguo Liao
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Mingyi Li
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
| | - Yi Liu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016 Liaoning China
| | - Rongrong He
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| | - Du Feng
- Key Laboratory of Protein Modification and Degradation, School of Basic Medical Sciences, Affiliated Cancer Hospital & Institute, Guangzhou, Medical University, Guangzhou, 511436 Guangdong China
| | - Runzhi Zhu
- Laboratory of Hepatobiliary Surgery, The Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001 Guangdong China
- Center for Cell Therapy, The Affiliated Hospital of Jiangsu University, Zhenjiang, 212001 Jiangsu China
| | - Hiroshi Kurihara
- College of Pharmacy, Jinan University, Guangzhou, 510632 Guangdong China
| |
Collapse
|
34
|
Gaseous signaling molecules and their application in resistant cancer treatment: from invisible to visible. Future Med Chem 2019; 11:323-336. [PMID: 30802141 DOI: 10.4155/fmc-2018-0403] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Multidrug resistance (MDR) in cancer remains a critical obstacle for efficient chemotherapy. Many MDR reversal agents have been discovered but failed in clinical trials due to severe toxic effects. Gaseous signaling molecules (GSMs), such as oxygen, nitric oxide, hydrogen sulfide and carbon monoxide, play key roles in regulating cell biological function and MDR. Compared with other toxic chemosensitizing agents, GSMs are endogenous and biocompatible molecules with little side effects. Research show that GSM modulators, including pharmaceutical formulations of GSMs (combined with conventional chemotherapeutic drugs) and GSM-donors (small molecules with GSMs releasing property), can overcome or reverse MDR. This review discusses the roles of these four GSMs in modulating MDR, and summarizes GSMs modulators in treating cancers with drug resistance.
Collapse
|
35
|
Zhao X, Cai A, Peng Z, Liang W, Xi H, Li P, Chen G, Yu J, Chen L. JS-K induces reactive oxygen species-dependent anti-cancer effects by targeting mitochondria respiratory chain complexes in gastric cancer. J Cell Mol Med 2019; 23:2489-2504. [PMID: 30672108 PMCID: PMC6433691 DOI: 10.1111/jcmm.14122] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 11/08/2018] [Accepted: 12/06/2018] [Indexed: 12/12/2022] Open
Abstract
As a nitric oxide (NO) donor prodrug, JS‐K inhibits cancer cell proliferation, induces the differentiation of human leukaemia cells, and triggers apoptotic cell death in various cancer models. However, the anti‐cancer effect of JS‐K in gastric cancer has not been reported. In this study, we found that JS‐K inhibited the proliferation of gastric cancer cells in vitro and in vivo and triggered mitochondrial apoptosis. Moreover, JS‐K induced a significant accumulation of reactive oxygen species (ROS), and the clearance of ROS by antioxidant reagents reversed JS‐K‐induced toxicity in gastric cancer cells and subcutaneous xenografts. Although JS‐K triggered significant NO release, NO scavenging had no effect on JS‐K‐induced toxicity in vivo and in vitro. Therefore, ROS, but not NO, mediated the anti‐cancer effects of JS‐K in gastric cancer. We also explored the potential mechanism of JS‐K‐induced ROS accumulation and found that JS‐K significantly down‐regulated the core proteins of mitochondria respiratory chain (MRC) complex I and IV, resulting in the reduction of MRC complex I and IV activity and the subsequent ROS production. Moreover, JS‐K inhibited the expression of antioxidant enzymes, including copper‐zinc‐containing superoxide dismutase (SOD1) and catalase, which contributed to the decrease of antioxidant enzymes activity and the subsequent inhibition of ROS clearance. Therefore, JS‐K may target MRC complex I and IV and antioxidant enzymes to exert ROS‐dependent anti‐cancer function, leading to the potential usage of JS‐K in the prevention and treatment of gastric cancer.
Collapse
Affiliation(s)
- Xudong Zhao
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Aizhen Cai
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zheng Peng
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Wenquan Liang
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hongqing Xi
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Peiyu Li
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| | - Guozhu Chen
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Jiyun Yu
- Institute of Military Cognitive and Brain Sciences, Academy of Military Medical Sciences, Beijing, China
| | - Lin Chen
- Department of General Surgery, Chinese People's Liberation Army General Hospital, Beijing, China
| |
Collapse
|
36
|
Chen L, Li X, Cheng M, Wang S, Zheng Q, Liu Q. Iso-pencillixanthone A from a marine-derived fungus reverses multidrug resistance in cervical cancer cells through down-regulating P-gp and re-activating apoptosis. RSC Adv 2018; 8:41192-41206. [PMID: 35559314 PMCID: PMC9091570 DOI: 10.1039/c8ra09506j] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2018] [Accepted: 11/30/2018] [Indexed: 12/24/2022] Open
Abstract
The occurrence of multidrug resistance (MDR) is highly associated with the overexpression of ATP-binding cassette (ABC) transporters, among which, P-glycoprotein (P-gp) plays one of the most important roles. Iso-pencillixanthone A (iso-PXA) is a compound isolated from the marine-derived fungus Penicillium oxalicum. No studies on the anti-tumor effect of this compound have been reported, except for a few focusing on its bactericidal properties. In this study, we found iso-PXA could stimulate P-gp ATPase activity and attenuate P-gp expression to increase the intracellular drug concentration in the cervical vincristine (VCR)-resistant cell line HeLa/VCR. Then, it increased ROS generation, depolarized MMP, promoted the release of cytochrome c from mitochondria, and further activated caspase-9, caspase-3 and PARP to induce cell apoptosis effectively through the intrinsic pathway. Caspase-8 medicated cleavage of Bid into the truncated form tBid partially initiated the mitochondrial apoptotic events. The elevation of the Bax/Bcl-2 ratio, the accumulation of FBW7 and the degradation of Mcl-1 accelerated the iso-PXA induced apoptotic process. The HeLa/VCR cell xenograft model again confirmed that iso-PXA had much better efficacy than vincristine in vivo. Taken together, these findings demonstrated that iso-PXA elicited remarkable anti-tumor and anti-MDR activity through inhibiting P-gp expression and function and re-activating the intrinsic apoptosis pathway in vitro and in vivo, suggesting it as a potential chemotherapeutic lead compound in the treatment of cervical MDR cancers.
Collapse
Affiliation(s)
- Li Chen
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University Fuzhou 350002 P. R. China
| | - Xinxin Li
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University Fuzhou 350002 P. R. China
| | - Miaomiao Cheng
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University Fuzhou 350002 P. R. China
| | - Siyuan Wang
- Institute of Biomedical and Pharmaceutical Technology, Fuzhou University Fuzhou 350002 P. R. China
| | - Qiuhong Zheng
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital Fuzhou 350014 P. R. China +86-591-8366-0063
| | - Qinying Liu
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fujian Cancer Hospital, Fujian Medical University Cancer Hospital Fuzhou 350014 P. R. China +86-591-8366-0063
| |
Collapse
|
37
|
Su X, Zhang L, Kang H, Zhang B, Bao G, Wang J. Mechanical, nanomorphological and biological reconstruction of early‑stage apoptosis in HeLa cells induced by cytochalasin B. Oncol Rep 2018; 41:928-938. [PMID: 30535459 PMCID: PMC6313055 DOI: 10.3892/or.2018.6921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2018] [Accepted: 11/22/2018] [Indexed: 12/29/2022] Open
Abstract
There is a growing interest in the fact that mechanical signals may be as important as biological signals in evaluating cell viability. To investigate the alterations in biomechanics, nanomorphology and biological apoptotic signals during early apoptosis, an apoptosis model was established for cervical cancer HeLa cells induced by cytochalasin B (CB). The cellular mechanical properties, geometry, morphology and expression of key apoptotic proteins were systematically analyzed. The findings indicated a marked decline in cellular elastic modulus and volume and a considerable increase in surface roughness occurring prior to the activation of biological apoptosis signals (such as phosphatidylserine exposure or activation of CD95/Fas). Moreover, the depolymerization of filamentous actin aggravated the intracellular crowding degree, which induced the redistribution of different-sized protein molecules and protrusions across the cell membrane arising from excluded volume interactions. Statistical analysis revealed that the disassembly of the actin cytoskeleton was negatively correlated with the cellular elastic modulus and volume, but was positively correlated with surface roughness and CD95/Fas activation. The results of the present study suggest that compared with biological signals, mechanical and geometrical reconstruction is more sensitive during apoptosis and the increase in cell surface roughness arises from the redistribution of biophysical molecules. These results contribute to our in-depth understanding of the apoptosis mechanisms of cancer cells mediated by cytochalasin B.
Collapse
Affiliation(s)
- Xuelian Su
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Ling Zhang
- Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Hong Kang
- Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Baoping Zhang
- Key Laboratory of Mechanics on Disaster and Environment in Western China, The Ministry of Education of China, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Guangjie Bao
- Key Laboratory of Stomatology of The State Ethnic Affairs Commission, Northwest Minzu University, Lanzhou, Gansu 730030, P.R. China
| | - Jizeng Wang
- College of Civil Engineering and Mechanics, Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|
38
|
Zhao XE, Yang Z, Zhang H, Yao G, Liu J, Wei Q, Ma B. Resveratrol Promotes Osteogenic Differentiation of Canine Bone Marrow Mesenchymal Stem Cells Through Wnt/Beta-Catenin Signaling Pathway. Cell Reprogram 2018; 20:371-381. [DOI: 10.1089/cell.2018.0032] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Affiliation(s)
- Xiao-e Zhao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Zhenshan Yang
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Hui Zhang
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Ge Yao
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Jie Liu
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Qiang Wei
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| | - Baohua Ma
- Key Laboratory of Animal Biotechnology, Ministry of Agriculture, Northwest A&F University, Yangling, China
- College of Veterinary Medicine, Northwest A&F University, Yangling, China
| |
Collapse
|
39
|
Qiu M, Zhang S, Ke L, Tang H, Zeng X, Liu J. JS-K enhances chemosensitivity of prostate cancer cells to Taxol via reactive oxygen species activation. Oncol Lett 2018; 17:757-764. [PMID: 30655827 PMCID: PMC6312932 DOI: 10.3892/ol.2018.9684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 05/15/2018] [Indexed: 12/24/2022] Open
Abstract
The aim of the present study was to investigate the influence of the nitric oxide donor prodrug JS-K (C13H16N6O8) on Taxol-induced apoptosis in prostate cancer cells, and to investigate a potential reactive oxygen species (ROS)-associated mechanism. The effect of JS-K on the anticancer activity of Taxol was assessed in prostate cancer cells; cell viability, colony formation, apoptosis, ROS generation and expression levels of apoptosis-associated proteins were investigated. The function of ROS accumulation in the combined effects of JS-K and Taxol was determined using the antioxidant N-acetylcysteine (NAC) and the pro-oxidant oxidized glutathione (GSSG). The results of the present study demonstrated that JS-K was able to increase Taxol-induced suppression of prostate cancer cell proliferation, apoptosis, ROS accumulation and upregulation of apoptosis-associated proteins. Furthermore, NAC reversed the effect of JS-K on Taxol-induced apoptosis and conversely, the pro-oxidant GSSG exacerbated the effect of JS-K on Taxol-induced apoptosis in prostate cancer cells. In conclusion, JS-K enhances the chemosensitivity of prostate cancer cells to Taxol, via the upregulation of intracellular ROS.
Collapse
Affiliation(s)
- Mingning Qiu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Sai Zhang
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Longzhi Ke
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Huancheng Tang
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Xin Zeng
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| | - Jianjun Liu
- Laboratory of Urology, Guangdong Medical University, Zhanjiang, Guangdong 524001, P.R. China
| |
Collapse
|
40
|
Dong SC, Sha HH, Xu XY, Hu TM, Lou R, Li H, Wu JZ, Dan C, Feng J. Glutathione S-transferase π: a potential role in antitumor therapy. Drug Des Devel Ther 2018; 12:3535-3547. [PMID: 30425455 PMCID: PMC6204874 DOI: 10.2147/dddt.s169833] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Glutathione S-transferase π (GSTπ) is a Phase II metabolic enzyme that is an important facilitator of cellular detoxification. Traditional dogma asserts that GSTπ functions to catalyze glutathione (GSH)-substrate conjunction to preserve the macromolecule upon exposure to oxidative stress, thus defending cells against various toxic compounds. Over the past 20 years, abnormal GSTπ expression has been linked to the occurrence of tumor resistance to chemotherapy drugs, demonstrating that this enzyme possesses functions beyond metabolism. This revelation reveals exciting possibilities in the realm of drug discovery, as GSTπ inhibitors and its prodrugs offer a feasible strategy in designing anticancer drugs with the primary purpose of reversing tumor resistance. In connection with the authors' current research, we provide a review on the biological function of GSTπ and current developments in GSTπ-targeting drugs, as well as the prospects of future strategies.
Collapse
Affiliation(s)
- Shu-Chen Dong
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huan-Huan Sha
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Xiao-Yue Xu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Tian-Mu Hu
- Department of Biological Science, Purdue University, West Lafayette, IN, USA
| | - Rui Lou
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Huizi Li
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jian-Zhong Wu
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Chen Dan
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| | - Jifeng Feng
- Jiangsu Cancer Hospital and Jiangsu Institute of Cancer Research and Nanjing Medical University Affiliated Cancer Hospital, Nanjing 210009, China, ;
| |
Collapse
|
41
|
Cheng C, Dong W. Aloe-Emodin Induces Endoplasmic Reticulum Stress-Dependent Apoptosis in Colorectal Cancer Cells. Med Sci Monit 2018; 24:6331-6339. [PMID: 30199885 PMCID: PMC6142869 DOI: 10.12659/msm.908400] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Recently, colorectal cancer has become a more common type of tumor in the world. Research has reported that several kinds of single compounds of Chinese herbs have shown anti-tumor activity in colorectal cancer. Aloe-emodin (AE), a natural compound extract from Aloe Vera, has been discovered to suppress cell proliferation and accelerate apoptosis in a variety of tumor cells. Whether AE exerts an effect on colorectal cancer cells has not yet been investigated. MATERIAL AND METHODS In this study, we examined the impact of AE on SW620 and HT29 colorectal cancer cell lines. After treatment with various concentrations of AE (10, 20, and 40 μM), cell proliferation, cell apoptosis, reactive oxygen species (ROS) generation, cytosolic calcium level, and related gene expression were analyzed. RESULTS Our results indicated that AE suppressed cell viability and induced cell apoptosis in SW620 and HT29 cell lines. Furthermore, both cell lines when exposed to AE generated ROS, which induces endoplasmic reticulum (ER) stress. We then detected the expression of ER stress-related proteins and cytosolic calcium levels. We found that cells exposure to AE had upregulation of unfolded protein response (UPR) proteins like glucose-related protein 78 (GRP78), phosphorylated protein kinase R (PKR)-like ER kinase (p-PERK), phosphorylated eukaryotic initiation factor-2α (p-eIF2α), and transcription factor C/EBP homologous protein (CHOP). Meanwhile, we detected an increased cytosolic calcium content followed by the upregulation of the calpain-1, calpain-2 and caspase-12. CHOP and caspase-12 are important regulatory factors leading to cell apoptosis. CONCLUSIONS AE might serve as a candidate in the treatment of colorectal cancer through inducing ER stress-dependent apoptosis.
Collapse
Affiliation(s)
- Chunsheng Cheng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China (mainland)
| |
Collapse
|
42
|
Huang Z, Liu L, Chen J, Cao M, Wang J. JS-K as a nitric oxide donor induces apoptosis via the ROS/Ca 2+/caspase-mediated mitochondrial pathway in HepG2 cells. Biomed Pharmacother 2018; 107:1385-1392. [PMID: 30257354 DOI: 10.1016/j.biopha.2018.08.142] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 08/16/2018] [Accepted: 08/25/2018] [Indexed: 02/07/2023] Open
Abstract
JS-K, (O2-(2, 4-dinitrophenyl) 1-[(4-ethoxycarbonyl) piperazin-1-yl] diazen 1-ium-1, 2-diolate), is a novel diazeniumdiolate-based nitric oxide donor prodrug. The present study investigated the relationship between reactive oxygen species (ROS) elevation, Ca2+ overload and mitochondrial disruption in JS-K-induced apoptosis. JS-K could significantly inhibit cell growth and induce apoptosis in a dose-dependent manner. Meanwhile, JS-K caused the accumulation of ROS, overload of Ca2+, decrease of mitochondrial membrane potential, release of cytochrome c (Cyt c) from mitochondria to the cytoplasm, increase of Bax-to-Bcl-2 ratio and activation of caspase- 9/3. NAC (an antioxidant) or BAPTA (an intracellular Ca2+ chelator) could partially reverse the above events, while BAPTA had little effect on the levels of ROS. Furthermore, pre-treatment with Carboxy-PTIO (a NO scavenger) significantly blocked the increasing of ROS, cytosolic Ca2+ and reversed the loss of mitochondrial membrane potential in JS-K-induced apoptosis. Taken together, the results suggested that NO released from JS-K could significantly induce HepG2 cell apoptosis through a ROS/Ca2+/caspase-mediated mitochondrial pathway.
Collapse
Affiliation(s)
- Zile Huang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| | - Ling Liu
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang 471003, Henan Province, China.
| | - Jingjing Chen
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| | - Mengyao Cao
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| | - Jiangang Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang 471003, Henan Province, China
| |
Collapse
|
43
|
Chen M, Du D, Zheng W, Liao M, Zhang L, Liang G, Gong M. Small hepatitis delta antigen selectively binds to target mRNA in hepatic cells: a potential mechanism by which hepatitis D virus downregulates glutathione S-transferase P1 and induces liver injury and hepatocarcinogenesis. Biochem Cell Biol 2018; 97:130-139. [PMID: 30153423 DOI: 10.1139/bcb-2017-0321] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Liver coinfection by hepatitis B virus (HBV) and hepatitis D virus (HDV) can result in a severe form of hepatocellular carcinoma with poor prognosis. Coinfection with HDV and HBV causes more deleterious effects than infection with HBV alone. Clinical research has shown that glutathione S-transferase P1 (GSTP1), a tumor suppressor gene, is typically downregulated in liver samples from hepatitis-infected patients. In the present study, our data indicated that small HDV antigen (s-HDAg) could specifically bind to GSTP1 mRNA and significantly downregulate GSTP1 protein expression. For the human fetal hepatocyte cell line L-02, cells transfected with s-HDAg, along with decreased GSTP1 expression, there was a significant accumulation of reactive oxygen species (ROS) and increased apoptotic ratios. Restoring GSTP1 expression through silencing s-HDAg via RNAi or overexpressing exogenous GSTP1 could largely recover the abnormal cell status. Our results revealed a novel potential mechanism of HDV-induced liver injury and hepatocarcinogenesis: s-HDAg can inhibit GSTP1 expression by directly binding to GSTP1 mRNA, which leads to accumulation of cellular ROS, resulting in high cellular apoptotic ratios and increased selective pressure for malignant transformation. To our knowledge, this is the first study to examine s-HDAg-specific pathogenic mechanisms through potential protein-RNA interactions.
Collapse
Affiliation(s)
- Mianzhi Chen
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Dan Du
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wen Zheng
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Mingheng Liao
- b Department of Liver Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lu Zhang
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ge Liang
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Meng Gong
- a Huaxi-Washington Mitochondria and Metabolism Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
44
|
Protein phosphatase 2A activation mechanism contributes to JS-K induced caspase-dependent apoptosis in human hepatocellular carcinoma cells. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2018; 37:142. [PMID: 29986744 PMCID: PMC6038275 DOI: 10.1186/s13046-018-0823-2] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Accepted: 07/03/2018] [Indexed: 12/11/2022]
Abstract
Background JS-K is a nitric oxide (NO) donor and could generate intracellularly high levels of NO. The study explores PP2A as a tumor suppressor is a major determinant mediating JS-K-caused apoptosis in human hepatocellular carcinoma (HCC) cells. Methods The human HCC cell lines (PLC5, Huh-7, Bel-7402, SMMC-7721 and HepG2) were used to assess effects of JS-K on cell viability, apoptosis induction and PP2A activation. Effects of JS-K on cell morphology, mitochondrial membrane potential, apoptosis and NO levels were determined in HCC cells expressing PP2A. Simultaneously, the expression of PP2A family including PP2A-A(α/β), PP2A-B55, PP2A-C(α/β) and the substrates of PP2A, such as β-catenin, c-Myc and p-Bcl-2 (Ser70) were detected in sensitive HCC cells. Furthermore, the role of NO in mediating the expression of PP2A was further validated with Z-VAD-FMK (a caspase inhibitor), Carboxy-PTIO (a NO scavenger), okadaic acid (OA, a PP2A inhibitor) and FTY720 (a PP2A agonist) in JS-K treated cells. In addition, the genetic manuplation of PP2A including overexpression and knockdown have been also performed in JS-K treated cells. Moreover, the rat model of primary hepatic carcinoma was established with diethylnitrosamine for 16 weeks to verify the anti-tumor effects of JS-K in vivo. Immunohistochemical and Western blot analysis were used to determine the expression of proteins in rat primary hepatic carcinoma tissues. Results JS-K significantly inhibited cell proliferation, increased apoptosis rate and activated PP2A activity in five HCC cells viability, especially SMMC7721 and HepG2 cells. It was characterized by loss of mitochondrial membrane potential, significant externalization of phosphatidylserine, nuclear morphological changes. Moreover, JS-K enhanced Bax-to-Bcl-2 ratio, released cytochrome c (Cyt c) from mitochondria, activated cleaved-caspase-9/3 and the cleavage of PARP, and decreased the expression of X-linked inhibitor of apoptosis protein (XIAP). Both Z-VAD-FMK and Carboxy-PTIO suppressed the activation of cleaved-caspase-9/3 and of cleaved-PARP in JS-K-treated sensitive HCC cells. Simultaneously, JS-K treatment could lead to the activation of protein phosphatase 2A-C (PP2A-C) but not PP2A-A and PP2A-B55, which subsequently inactivated and dephosphorylated the PP2A substrates including β-catenin, c-Myc, and p-Bcl-2 (Ser70). However, silencing PP2A-C could abolish both the activation of PP2A-C and down-regulation of β-catenin, c-Myc and p-Bcl-2 (Ser70) in sensitive HCC cells. Conversely, PP2A overexpression could enhance the effects of JS-K on activation of PP2A and down-regulation of β-catenin, c-Myc and p-Bcl-2 (Ser70). In addition, adding okadaic acid (OA), a PP2A inhibitor, abolished the effects of JS-K on apoptosis induction, PP2A activation and the substrates of PP2A dephosphorylation; FTY720, a PP2A agonist, enhanced the effects of JS-K including apoptosis induction, PP2A activation and the substrates of PP2A dephosphorylation. The mice exhibited a lower number and smaller tumor nodules in response to JS-K-treated group. A marked increase in the number of hepatocytes with PCNA-positive nuclei (proliferating cells) was evident in DEN group and tended to decrease with JS-K treatment. Furthermore, JS-K treatment could induce PP2A activation and the substrates of PP2A inactivation such as β-catenin, c-Myc and p-Bcl-2(Ser70) in DEN-induced hepatocarcinogenesis. Conclusions High levels of NO released from JS-K induces a caspase-dependent apoptosis through PP2A activation.
Collapse
|
45
|
Liu L, Huang Z, Chen J, Wang J, Wang S. Protein phosphatase 2A mediates JS-K-induced apoptosis by affecting Bcl-2 family proteins in human hepatocellular carcinoma HepG2 cells. J Cell Biochem 2018; 119:6633-6643. [PMID: 29693750 DOI: 10.1002/jcb.26845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Accepted: 03/09/2018] [Indexed: 12/18/2022]
Abstract
Protein phosphatase 2A (PP2A) is an important enzyme within various signal transduction pathways. The present study was investigated PP2A mediates JS-K-induced apoptosis by affecting Bcl-2 family protein. JS-K showed diverse inhibitory effects in five HCC cell lines, especially HepG2 cells. JS-K caused a dose- and time-dependent reduction in cell viability and increased in levels of LDH release. Meanwhile, JS-K- induced apoptosis was characterized by mitochondrial membrane potential reduction, Hoechst 33342+ /PI+ dual staining, release of cytochrome c (Cyt c), and activation of cleaved caspase-9/3. Moreover, JS-K-treatment could lead to the activation of protein phosphatase 2A-C (PP2A-C), decrease of anti-apoptotic Bcl-2 family-protein expression including p-Bcl-2 (Ser70), Bcl-2, Bcl-xL, and Mcl-1 as well as the increase of pro-apoptosis Bcl-2 family-protein including Bim, Bad, Bax, and Bak. Furthermore, JS-K caused a marked increase of intracellular NO levels while pre-treatment with Carboxy-PTIO (a NO scavenger) reduced the cytotoxicity effects and the apoptosis rate. Meanwhile, pre-treatment with Carboxy-PTIO attenuated the JS-K-induced up-regulation of PP2A, Cyt c, and cleaved-caspase-9/3 activation. The silencing PP2A-C by siRNA could abolish the activation of PP2A-C, down-regulation of anti-apoptotic Bcl-2 family-protein (p-Bcl-2, Bcl-2, Bcl-xL, and Mcl-1), increase of pro-apoptosis Bcl-2 family-protein (Bim, Bad, Bax, and Bak) and apoptotic-related protein (Cyt c, cleaved caspase-9/3) that were caused by JS-K in HepG2 cells. In addition, pre-treatment with OA (a PP2A inhibitor) also attenuated the above effects induced by JS-K. In summary, NO release from JS-K induces apoptosis through PP2A activation, which contributed to the regulation of Bcl-2 family proteins.
Collapse
Affiliation(s)
- Ling Liu
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Zile Huang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jingjing Chen
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Jiangang Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| | - Shuying Wang
- Department of Pharmacy, Medical College, Henan University of Science and Technology, Luoyang, China
| |
Collapse
|
46
|
Study on the effect of reactive oxygen species-mediated oxidative stress on the activation of mitochondrial apoptosis and the tenderness of yak meat. Food Chem 2018; 244:394-402. [DOI: 10.1016/j.foodchem.2017.10.034] [Citation(s) in RCA: 74] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2017] [Revised: 09/16/2017] [Accepted: 10/09/2017] [Indexed: 12/17/2022]
|
47
|
Naiki T, Naiki-Ito A, Iida K, Etani T, Kato H, Suzuki S, Yamashita Y, Kawai N, Yasui T, Takahashi S. GPX2 promotes development of bladder cancer with squamous cell differentiation through the control of apoptosis. Oncotarget 2018; 9:15847-15859. [PMID: 29662611 PMCID: PMC5882302 DOI: 10.18632/oncotarget.24627] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2017] [Accepted: 02/27/2018] [Indexed: 01/10/2023] Open
Abstract
Herein, we elucidated the molecular mechanisms and therapeutic potential of glutathione peroxidase 2 (GPX2) in bladder cancer. GPX2 expression gradually increased during progression from normal to papillary or nodular hyperplasia (PNHP) and urothelial carcinoma (UC) in a rat N-butyl-N-(4-hydroxybutyl) nitrosamine (BBN)-induced bladder carcinogenesis model. GPX2 overexpression was more marked in UC with squamous differentiation (SqD) than in pure UC. Clinical intraepithelial lesions of papillary UC and invasive UC with SqD also had strong GPX2 expression in human radical cystectomy specimens. In addition, prognostic analysis using transurethral specimens revealed that low expression level of GPX2 predicted poor prognosis in patients with pure UC. Further, UC cell lines, BC31 and RT4, cultured in vitro also overexpressed GPX2. Knock-down of GPX2 induced significant inhibition of intracellular reactive oxygen species (ROS) production, in addition to significant growth inhibition and increased apoptosis with activation of caspase 3 or 7 in both BC31 and RT4 cells. Interestingly, tumor growth of BC31 cells subcutaneously transplanted in nude mice was significantly caused the induction of apoptosis, as well as inhibition of angiogenesis and SqD by GPX2 down-regulation. Our findings demonstrated that GPX2 plays an important role in bladder carcinogenesis through the regulation of apoptosis against intracellular ROS, and may be considered as a novel biomarker or therapeutic target in bladder cancer.
Collapse
Affiliation(s)
- Taku Naiki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan.,Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Aya Naiki-Ito
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Keitaro Iida
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Toshiki Etani
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Hiroyuki Kato
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Shugo Suzuki
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Yoriko Yamashita
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Noriyasu Kawai
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Takahiro Yasui
- Department of Nephro-Urology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| | - Satoru Takahashi
- Department of Experimental Pathology and Tumor Biology, Nagoya City University, Graduate School of Medical Sciences, Nagoya, Japan
| |
Collapse
|
48
|
Reactive oxygen species stress increases accumulation of tyrosyl-DNA phsosphodiesterase 1 within mitochondria. Sci Rep 2018. [PMID: 29523818 PMCID: PMC5844879 DOI: 10.1038/s41598-018-22547-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Tyrosyl-DNA phosphodiesterase 1 (Tdp1) is a nuclear and mitochondrial protein that in nuclei and in vitro repairs blocked 3' DNA termini such as 3' phosphotyrosine conjugates resulting from stalling of topoisomerase I-DNA intermediates. Its mutation also causes spinocerebellar ataxia with axonal neuropathy type 1 (SCAN1). Because Tdp1 colocalizes with mitochondria following oxidative stress, we hypothesized that Tdp1 repairs mitochondrial DNA (mtDNA) and that mtDNA damage mediates entry of Tdp1 into the mitochondria. To test this, we used S. cerevisiae mutants, cultured mouse and human cells, and a Tdp1 knockout mouse. H2O2- and rotenone-induced cellular and intramitochondrial reactive oxygen species (ROS) activated oxidant-responsive kinases P38 and ERK1, and the translocation of Tdp1 from the nucleus to the mitochondria via the TIM/TOM complex. This translocation occurred independently of mtDNA. Within the mitochondria, Tdp1 interacted with Ligase III and reduced mtDNA mutations. Tdp1-deficient tissues had impaired mitochondrial respiration and decreased viability. These observations suggest that Tdp1 maintains mtDNA integrity and support the hypothesis that mitochondrial dysfunction contributes to the pathology of SCAN1.
Collapse
|
49
|
Zhang J, Song H, Ji S, Wang X, Huang P, Zhang C, Wang W, Kong D. NO prodrug-conjugated, self-assembled, pH-responsive and galactose receptor targeted nanoparticles for co-delivery of nitric oxide and doxorubicin. NANOSCALE 2018; 10:4179-4188. [PMID: 29442103 DOI: 10.1039/c7nr08176f] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Targeted delivery and controlled release of nitric oxide (NO) locoregionally are in high demand and challenging in cancer treatment. Herein, we report an example of galactose receptor targeted, pH-responsive and self-assembled nanoparticle-based delivery of the NO prodrug O2-(2,4-dinitrophenyl) 1-[4-(propargyloxycarbonyl)piperazin-1-yl]diazen-1-ium-1,2-diolate (alkynyl-JSK), which was chemically conjugated to an amphiphilic block copolymer through a click reaction for the first time. The assembled NO prodrug nanoparticles show high NO capacity (the content of the NO prodrug in the copolymer, ∼23.4% (w/w)), good stability and a sustained NO release pattern with unique glutathione/glutathione S-transferase (GSH/GST) activated NO-releasing kinetics. Such NO-loaded nanoparticles exhibit superior cytotoxicity to HepG2 cells. More importantly, in combination with doxorubicin (DOX) chemotherapy a significant synergistic therapeutic effect was achieved, due to its excellent galactose receptor-targeting capability, rapid acid-triggered DOX release and sustained NO release. Our findings indicate that these multifunctional nanoparticles can serve as an efficient NO and chemotherapeutic agent delivery platform, holding great promise in cancer combinatorial treatment.
Collapse
Affiliation(s)
- Jimin Zhang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Collaborative Innovation Center of Chemical Science and Engineering (Tianjin), College of Life Sciences, Nankai University, Tianjin 300071, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
50
|
Qiu M, Shi F, Dai F, Song R, Wang S, You Y, Zhao B. A reactive oxygen species activation mechanism contributes to Sophoridine-induced apoptosis in rat liver BRL-3A cells. JOURNAL OF ETHNOPHARMACOLOGY 2018; 213:376-383. [PMID: 29102763 DOI: 10.1016/j.jep.2017.10.030] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Revised: 10/16/2017] [Accepted: 10/29/2017] [Indexed: 06/07/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Sophora alopecuroides L., a traditional Chinese herb, has been widely used to treat numerous diseases throughout China. Quinolizidine alkaloids were identified as active components in Sophora alopecuroides L., and Sophoridine (SRI) is the major component in the Quinolizidine alkaloids. AIM OF THE STUDY To investigate the toxic effects of SRI in rat liver BRL-3A cells and to explore potential ROS-related mechanisms. MATERIALS AND METHODS Cell viability, cytotoxicity, apoptosis, intracellular generation of ROS, GSH/GSSG ratio and levels of proteins in mitochondria apoptosis pathway were analyzed. RESULTS Our data indicated that SRI could suppress BRL-3A cells viability in a concentration- and time-dependent manner and increase cytotoxicity, ROS accumulation and cell apoptosis in a concentration-dependent manner. Expressions and activities of apoptotic related proteins were upregulated, whereas expression of Bcl-2 was downregulated after treatment. Furthermore, level of H2O2 was increased, whereas level of Superoxide was not changed after treatment. Moreover, the antioxidant N-acetylcysteine reversed SRI-induced apoptosis and ROS accumulation. CONCLUSION Our data suggest that SRI promotes rat liver BRL-3A cells apoptosis by increasing intracellular ROS accumulation.
Collapse
Affiliation(s)
- Mingning Qiu
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fangyun Shi
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Fei Dai
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Runjie Song
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Shuai Wang
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Yanfei You
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China
| | - Baoyu Zhao
- College of Veterinary Medicine, Northwest A&F University, Yangling, Shaanxi 712100, China; Institute of Poisonous Plants in Western China, Northwest A&F University, Yangling, Shaanxi 712100, China.
| |
Collapse
|